401
|
Ignatieva EV, Afonnikov DA, Saik OV, Rogaev EI, Kolchanov NA. A compendium of human genes regulating feeding behavior and body weight, its functional characterization and identification of GWAS genes involved in brain-specific PPI network. BMC Genet 2016; 17:158. [PMID: 28105929 PMCID: PMC5249002 DOI: 10.1186/s12863-016-0466-2] [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] [Indexed: 12/31/2022] Open
Abstract
Background Obesity is heritable. It predisposes to many diseases. The objectives of this study were to create a compendium of genes relevant to feeding behavior (FB) and/or body weight (BW) regulation; to construct and to analyze networks formed by associations between genes/proteins; and to identify the most significant genes, biological processes/pathways, and tissues/organs involved in BW regulation. Results The compendium of genes controlling FB or BW includes 578 human genes. Candidate genes were identified from various sources, including previously published original research and review articles, GWAS meta-analyses, and OMIM (Online Mendelian Inheritance in Man). All genes were ranked according to knowledge about their biological role in body weight regulation and classified according to expression patterns or functional characteristics. Substantial and overrepresented numbers of genes from the compendium encoded cell surface receptors, signaling molecules (hormones, neuropeptides, cytokines), transcription factors, signal transduction proteins, cilium and BBSome components, and lipid binding proteins or were present in the brain-specific list of tissue-enriched genes identified with TSEA tool. We identified 27 pathways from KEGG, REACTOME and BIOCARTA whose genes were overrepresented in the compendium. Networks formed by physical interactions or homological relationships between proteins or interactions between proteins involved in biochemical/signaling pathways were reconstructed and analyzed. Subnetworks and clusters identified by the MCODE tool included genes/proteins associated with cilium morphogenesis, signal transduction proteins (particularly, G protein–coupled receptors, kinases or proteins involved in response to insulin stimulus) and transcription regulation (particularly nuclear receptors). We ranked GWAS genes according to the number of neighbors in three networks and revealed 22 GWAS genes involved in the brain-specific PPI network. On the base of the most reliable PPIs functioning in the brain tissue, new regulatory schemes interpreting relevance to BW regulation are proposed for three GWAS genes (ETV5, LRP1B, and NDUFS3). Conclusions A compendium comprising 578 human genes controlling FB or BW was designed, and the most significant functional groups of genes, biological processes/pathways, and tissues/organs involved in BW regulation were revealed. We ranked genes from the GWAS meta-analysis set according to the number and quality of associations in the networks and then according to their involvement in the brain-specific PPI network and proposed new regulatory schemes involving three GWAS genes (ETV5, LRP1B, and NDUFS3) in BW regulation. The compendium is expected to be useful for pathology risk estimation and for design of new pharmacological approaches in the treatment of human obesity. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0466-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena V Ignatieva
- Center for Brain Neurobiology and Neurogenetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia. .,Laboratory of Evolutionary Bioinformatics and Theoretical Genetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Dmitry A Afonnikov
- Center for Brain Neurobiology and Neurogenetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia.,Laboratory of Evolutionary Bioinformatics and Theoretical Genetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Olga V Saik
- Center for Brain Neurobiology and Neurogenetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Evgeny I Rogaev
- Center for Brain Neurobiology and Neurogenetics, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.,BNRI, Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, 15604, USA
| | - Nikolay A Kolchanov
- Novosibirsk State University, Novosibirsk, 630090, Russia.,Department of Systems Biology, The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
402
|
Xie H, Guo R, Zhong H, Feng Q, Lan Z, Qin B, Ward KJ, Jackson MA, Xia Y, Chen X, Chen B, Xia H, Xu C, Li F, Xu X, Al-Aama JY, Yang H, Wang J, Kristiansen K, Wang J, Steves CJ, Bell JT, Li J, Spector TD, Jia H. Shotgun Metagenomics of 250 Adult Twins Reveals Genetic and Environmental Impacts on the Gut Microbiome. Cell Syst 2016; 3:572-584.e3. [PMID: 27818083 PMCID: PMC6309625 DOI: 10.1016/j.cels.2016.10.004] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/13/2016] [Accepted: 10/05/2016] [Indexed: 12/14/2022]
Abstract
The gut microbiota has been typically viewed as an environmental factor for human health. Twins are well suited for investigating the concordance of their gut microbiomes and decomposing genetic and environmental influences. However, existing twin studies utilizing metagenomic shotgun sequencing have included only a few samples. Here, we sequenced fecal samples from 250 adult twins in the TwinsUK registry and constructed a comprehensive gut microbial reference gene catalog. We demonstrate heritability of many microbial taxa and functional modules in the gut microbiome, including those associated with diseases. Moreover, we identified 8 million SNPs in the gut microbiome and observe a high similarity in microbiome SNPs between twins that slowly decreases after decades of living apart. The results shed new light on the genetic and environmental influences on the composition and function of the gut microbiome that could relate to risk of complex diseases.
Collapse
Affiliation(s)
- Hailiang Xie
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Ruijin Guo
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China; Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Huanzi Zhong
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Qiang Feng
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China
| | - Zhou Lan
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Kirsten J Ward
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Matthew A Jackson
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Yan Xia
- BGI-Shenzhen, Shenzhen 518083, China; BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Xu Chen
- BGI-Shenzhen, Shenzhen 518083, China; Qingdao University-BGI Joint Innovation College, Qingdao University, Qingdao 266071, China
| | - Bing Chen
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Huihua Xia
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China
| | - Changlu Xu
- BGI-Shenzhen, Shenzhen 518083, China; Qingdao University-BGI Joint Innovation College, Qingdao University, Qingdao 266071, China
| | - Fei Li
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | | | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen 518083, China; Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Jun Wang
- BGI-Shenzhen, Shenzhen 518083, China; Macau University of Science and Technology, Taipa, Macau 999078, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Junhua Li
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China.
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK.
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen 518083, China; China National Genebank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China; Macau University of Science and Technology, Taipa, Macau 999078, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China.
| |
Collapse
|
403
|
Wang Z, Sun Y, Fu X, Yu G, Wang C, Bao F, Yue Z, Li J, Sun L, Irwanto A, Yu Y, Chen M, Mi Z, Wang H, Huai P, Li Y, Du T, Yu W, Xia Y, Xiao H, You J, Li J, Yang Q, Wang N, Shang P, Niu G, Chi X, Wang X, Cao J, Cheng X, Liu H, Liu J, Zhang F. A large-scale genome-wide association and meta-analysis identified four novel susceptibility loci for leprosy. Nat Commun 2016; 7:13760. [PMID: 27976721 PMCID: PMC5172377 DOI: 10.1038/ncomms13760] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/31/2016] [Indexed: 11/18/2022] Open
Abstract
Leprosy, a chronic infectious disease, results from the uncultivable pathogen Mycobacterium leprae (M. leprae), and usually progresses to peripheral neuropathy and permanent progressive deformity if not treated. Previously published genetic studies have identified 18 gene/loci significantly associated with leprosy at the genome-wide significant level. However as a complex disease, only a small proportion of leprosy risk could be explained by those gene/loci. To further identify more susceptibility gene/loci, we hereby performed a three-stage GWAS comprising 8,156 leprosy patients and 15,610 controls of Chinese ancestry. Four novel loci were identified including rs6807915 on 3p25.2 (P=1.94 × 10−8, OR=0.89), rs4720118 on 7p14.3 (P=3.85 × 10−10, OR=1.16), rs55894533 on 8p23.1 (P=5.07 × 10−11, OR=1.15) and rs10100465 on 8q24.11 (P=2.85 × 10−11, OR=0.85). Altogether, these findings have provided new insight and significantly expanded our understanding of the genetic basis of leprosy.
Previous studies have shown genetic associations between leprosy and 18 different genes/loci. Here, Wang and colleagues perform genome-wide association study in Han Chinese leprosy patients and describe four novel loci to be associated to the disease.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Yonghu Sun
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Xi'an Fu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Gongqi Yu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine and Life Science, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong 250022, China
| | - Chuan Wang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Fangfang Bao
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Zhenhua Yue
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Jianke Li
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Lele Sun
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Astrid Irwanto
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Yongxiang Yu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Mingfei Chen
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Zihao Mi
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Honglei Wang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Pengcheng Huai
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Yi Li
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Tiantian Du
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Wenjun Yu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Yang Xia
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Hailu Xiao
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Jiabao You
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Jinghui Li
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Qing Yang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Na Wang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Panpan Shang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Guiye Niu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China
| | - Xiaojun Chi
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Xiuhuan Wang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Jing Cao
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Xiujun Cheng
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China
| | - Hong Liu
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China
| | - Jianjun Liu
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Furen Zhang
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.,Shandong Provincial Key Laboratory for Dermatovenereology, Jinan, Shandong 250000, China.,School of Medicine, Shandong University, Jinan, Shandong 250000, China.,School of Medicine and Life Science, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong 250022, China.,Shandong Provincial Hospital for Skin Diseases, Shandong University, Jinan, Shandong 250000, China.,National Clinical Key Project of Dermatology and Venereology, Jinan, Shandong 250000, China
| |
Collapse
|
404
|
Keller MP, Paul PK, Rabaglia ME, Stapleton DS, Schueler KL, Broman AT, Ye SI, Leng N, Brandon CJ, Neto EC, Plaisier CL, Simonett SP, Kebede MA, Sheynkman GM, Klein MA, Baliga NS, Smith LM, Broman KW, Yandell BS, Kendziorski C, Attie AD. The Transcription Factor Nfatc2 Regulates β-Cell Proliferation and Genes Associated with Type 2 Diabetes in Mouse and Human Islets. PLoS Genet 2016; 12:e1006466. [PMID: 27935966 PMCID: PMC5147809 DOI: 10.1371/journal.pgen.1006466] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/04/2016] [Indexed: 12/22/2022] Open
Abstract
Human genome-wide association studies (GWAS) have shown that genetic variation at >130 gene loci is associated with type 2 diabetes (T2D). We asked if the expression of the candidate T2D-associated genes within these loci is regulated by a common locus in pancreatic islets. Using an obese F2 mouse intercross segregating for T2D, we show that the expression of ~40% of the T2D-associated genes is linked to a broad region on mouse chromosome (Chr) 2. As all but 9 of these genes are not physically located on Chr 2, linkage to Chr 2 suggests a genomic factor(s) located on Chr 2 regulates their expression in trans. The transcription factor Nfatc2 is physically located on Chr 2 and its expression demonstrates cis linkage; i.e., its expression maps to itself. When conditioned on the expression of Nfatc2, linkage for the T2D-associated genes was greatly diminished, supporting Nfatc2 as a driver of their expression. Plasma insulin also showed linkage to the same broad region on Chr 2. Overexpression of a constitutively active (ca) form of Nfatc2 induced β-cell proliferation in mouse and human islets, and transcriptionally regulated more than half of the T2D-associated genes. Overexpression of either ca-Nfatc2 or ca-Nfatc1 in mouse islets enhanced insulin secretion, whereas only ca-Nfatc2 was able to promote β-cell proliferation, suggesting distinct molecular pathways mediating insulin secretion vs. β-cell proliferation are regulated by NFAT. Our results suggest that many of the T2D-associated genes are downstream transcriptional targets of NFAT, and may act coordinately in a pathway through which NFAT regulates β-cell proliferation in both mouse and human islets. Genome-wide association studies (GWAS) and linkage studies provide a powerful way to establish a causal connection between a gene locus and a physiological or pathophysiological phenotype. We wondered if candidate genes associated with type 2 diabetes in human populations, in addition to being causal for the disease, could also be intermediate traits in a pathway leading to disease. In addition, we wished to know if there were any regulatory loci that could coordinately drive the expression of these genes in pancreatic islets and thus complete a pathway; i.e. Driver → GWAS candidate expression → type 2 diabetes. Using data from a mouse intercross between a diabetes-susceptible and a diabetes-resistant mouse strain, we found that the expression of ~40% of >130 candidate GWAS genes genetically mapped to a hot spot on mouse chromosome 2. Using a variety of statistical methods, we identified the transcription factor Nfatc2 as the candidate driver. Follow-up experiments showed that overexpression of Nfatc2 does indeed affect the expression of the GWAS genes and regulates β-cell proliferation and insulin secretion. The work shows that in addition to being causal, GWAS candidate genes can be intermediate traits in a pathway leading to disease. Model organisms can be used to explore these novel causal pathways.
Collapse
Affiliation(s)
- Mark P. Keller
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Pradyut K. Paul
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Mary E. Rabaglia
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Donnie S. Stapleton
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kathryn L. Schueler
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Aimee Teo Broman
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Shuyun Isabella Ye
- Department of Statistics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ning Leng
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Christopher J. Brandon
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | | | | | - Shane P. Simonett
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Melkam A. Kebede
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Gloria M. Sheynkman
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Mark A. Klein
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | | | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Karl W. Broman
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Brian S. Yandell
- Department of Statistics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Christina Kendziorski
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
405
|
Corella D, Coltell O, Sorlí JV, Estruch R, Quiles L, Martínez-González MÁ, Salas-Salvadó J, Castañer O, Arós F, Ortega-Calvo M, Serra-Majem L, Gómez-Gracia E, Portolés O, Fiol M, Díez Espino J, Basora J, Fitó M, Ros E, Ordovás JM. Polymorphism of the Transcription Factor 7-Like 2 Gene (TCF7L2) Interacts with Obesity on Type-2 Diabetes in the PREDIMED Study Emphasizing the Heterogeneity of Genetic Variants in Type-2 Diabetes Risk Prediction: Time for Obesity-Specific Genetic Risk Scores. Nutrients 2016; 8:793. [PMID: 27929407 PMCID: PMC5188448 DOI: 10.3390/nu8120793] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/17/2016] [Accepted: 11/17/2016] [Indexed: 11/24/2022] Open
Abstract
Nutrigenetic studies analyzing gene-diet interactions of the TCF7L2-rs7903146 C > T polymorphism on type-2 diabetes (T2D) have shown controversial results. A reason contributing to this may be the additional modulation by obesity. Moreover, TCF7L2-rs7903146 is one of the most influential variants in T2D-genetic risk scores (GRS). Therefore, to increase the predictive value (PV) of GRS it is necessary to first see whether the included polymorphisms have heterogeneous effects. We comprehensively investigated gene-obesity interactions between the TCF7L2-rs7903146 C > T polymorphism on T2D (prevalence and incidence) and analyzed other T2D-polymorphisms in a sub-sample. We studied 7018 PREDIMED participants at baseline and longitudinally (8.7 years maximum follow-up). Obesity significantly interacted with the TCF7L2-rs7903146 on T2D prevalence, associations being greater in non-obese subjects. Accordingly, we prospectively observed in non-T2D subjects (n = 3607) that its association with T2D incidence was stronger in non-obese (HR: 1.81; 95% CI: 1.13-2.92, p = 0.013 for TT versus CC) than in obese subjects (HR: 1.01; 95% CI: 0.61-1.66; p = 0.979; p-interaction = 0.048). Accordingly, TCF7L2-PV was higher in non-obese subjects. Additionally, we created obesity-specific GRS with ten T2D-polymorphisms and demonstrated for the first time their higher strata-specific PV. In conclusion, we provide strong evidence supporting the need for considering obesity when analyzing the TCF7L2 effects and propose the use of obesity-specific GRS for T2D.
Collapse
Affiliation(s)
- Dolores Corella
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Oscar Coltell
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Computer Languages and Systems, School of Technology and Experimental Sciences, Universitat Jaume I, 12071 Castellón, Spain.
| | - Jose V Sorlí
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Ramón Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Internal Medicine, Hospital Clinic, IDIBAPS, 08036 Barcelona, Spain.
| | - Laura Quiles
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Miguel Ángel Martínez-González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Preventive Medicine and Public Health, University of Navarra-Navarra Institute for Health Research (IdisNa), 31009 Pamplona, Spain.
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Human Nutrition Unit, Biochemistry and Biotechnology Department, IISPV, University Rovira i Virgili, 43003 Reus, Spain.
| | - Olga Castañer
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain.
| | - Fernando Arós
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Cardiology, Hospital Txagorritxu, 01009 Vitoria, Spain.
| | - Manuel Ortega-Calvo
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Family Medicine, Distrito Sanitario Atención Primaria Sevilla, Centro de Salud Las Palmeritas, 41003 Sevilla, Spain.
| | - Lluís Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, 35001 Las Palmas de Gran Canaria, Spain.
| | - Enrique Gómez-Gracia
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Epidemiology, School of Medicine, University of Malaga, 29071 Malaga, Spain.
| | - Olga Portolés
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Miquel Fiol
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Palma Institute of Health Research (IdISPa), Hospital Son Espases, 07014 Palma de Mallorca, Spain.
| | - Javier Díez Espino
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Preventive Medicine and Public Health, University of Navarra-Navarra Institute for Health Research (IdisNA)-Servicio Navarro de Salud-Osasunbidea, 31009 Pamplona, Spain.
| | - Josep Basora
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Human Nutrition Unit, Biochemistry and Biotechnology Department, IISPV, University Rovira i Virgili, 43003 Reus, Spain.
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain.
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, 08036 Barcelona, Spain.
| | - José M Ordovás
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA.
- Department of Cardiovascular Epidemiology and Population Genetics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029-IMDEA Alimentación, 28049 Madrid, Spain.
| |
Collapse
|
406
|
Multiethnic genome-wide meta-analysis of ectopic fat depots identifies loci associated with adipocyte development and differentiation. Nat Genet 2016; 49:125-130. [PMID: 27918534 DOI: 10.1038/ng.3738] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 11/07/2016] [Indexed: 02/08/2023]
Abstract
Variation in body fat distribution contributes to the metabolic sequelae of obesity. The genetic determinants of body fat distribution are poorly understood. The goal of this study was to gain new insights into the underlying genetics of body fat distribution by conducting sample-size-weighted fixed-effects genome-wide association meta-analyses in up to 9,594 women and 8,738 men of European, African, Hispanic and Chinese ancestry, with and without sex stratification, for six traits associated with ectopic fat (hereinafter referred to as ectopic-fat traits). In total, we identified seven new loci associated with ectopic-fat traits (ATXN1, UBE2E2, EBF1, RREB1, GSDMB, GRAMD3 and ENSA; P < 5 × 10-8; false discovery rate < 1%). Functional analysis of these genes showed that loss of function of either Atxn1 or Ube2e2 in primary mouse adipose progenitor cells impaired adipocyte differentiation, suggesting physiological roles for ATXN1 and UBE2E2 in adipogenesis. Future studies are necessary to further explore the mechanisms by which these genes affect adipocyte biology and how their perturbations contribute to systemic metabolic disease.
Collapse
|
407
|
Chen BH, Hivert MF, Peters MJ, Pilling LC, Hogan JD, Pham LM, Harries LW, Fox CS, Bandinelli S, Dehghan A, Hernandez DG, Hofman A, Hong J, Joehanes R, Johnson AD, Munson PJ, Rybin DV, Singleton AB, Uitterlinden AG, Ying S, Melzer D, Levy D, van Meurs JBJ, Ferrucci L, Florez JC, Dupuis J, Meigs JB, Kolaczyk ED. Peripheral Blood Transcriptomic Signatures of Fasting Glucose and Insulin Concentrations. Diabetes 2016; 65:3794-3804. [PMID: 27625022 PMCID: PMC5127245 DOI: 10.2337/db16-0470] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/04/2016] [Indexed: 01/09/2023]
Abstract
Genome-wide association studies (GWAS) have successfully identified genetic loci associated with glycemic traits. However, characterizing the functional significance of these loci has proven challenging. We sought to gain insights into the regulation of fasting insulin and fasting glucose through the use of gene expression microarray data from peripheral blood samples of participants without diabetes in the Framingham Heart Study (FHS) (n = 5,056), the Rotterdam Study (RS) (n = 723), and the InCHIANTI Study (Invecchiare in Chianti) (n = 595). Using a false discovery rate q <0.05, we identified three transcripts associated with fasting glucose and 433 transcripts associated with fasting insulin levels after adjusting for age, sex, technical covariates, and complete blood cell counts. Among the findings, circulating IGF2BP2 transcript levels were positively associated with fasting insulin in both the FHS and RS. Using 1000 Genomes-imputed genotype data, we identified 47,587 cis-expression quantitative trait loci (eQTL) and 6,695 trans-eQTL associated with the 433 significant insulin-associated transcripts. Of note, we identified a trans-eQTL (rs592423), where the A allele was associated with higher IGF2BP2 levels and with fasting insulin in an independent genetic meta-analysis comprised of 50,823 individuals. We conclude that integration of genomic and transcriptomic data implicate circulating IGF2BP2 mRNA levels associated with glucose and insulin homeostasis.
Collapse
Affiliation(s)
- Brian H Chen
- Longitudinal Studies Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA
- Diabetes Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Marjolein J Peters
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
| | - Luke C Pilling
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - John D Hogan
- Program in Bioinformatics, Boston University, Boston, MA
| | - Lisa M Pham
- Program in Bioinformatics, Boston University, Boston, MA
| | - Lorna W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Caroline S Fox
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Stefania Bandinelli
- Geriatric Rehabilitation Unit, Azienda Sanitaria di Firenze, Florence, Italy
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dena G Hernandez
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Roby Joehanes
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
- Hebrew SeniorLife, Harvard Medical School, Boston, MA
| | - Andrew D Johnson
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Peter J Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD
| | - Denis V Rybin
- Data Coordinating Center, Boston University, Boston, MA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Saixia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD
| | | | - David Melzer
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
- Netherlands Genomics Initiative-sponsored Netherlands Consortium for Healthy Aging, Leiden and Rotterdam, the Netherlands
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Jose C Florez
- Diabetes Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA
- Metabolism Program and Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Josée Dupuis
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - James B Meigs
- Metabolism Program and Program in Medical and Population Genetics, Broad Institute, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric D Kolaczyk
- Program in Bioinformatics, Boston University, Boston, MA
- Department of Mathematics and Statistics, Boston University, MA
| |
Collapse
|
408
|
Snijders AM, Langley SA, Kim YM, Brislawn CJ, Noecker C, Zink EM, Fansler SJ, Casey CP, Miller DR, Huang Y, Karpen GH, Celniker SE, Brown JB, Borenstein E, Jansson JK, Metz TO, Mao JH. Influence of early life exposure, host genetics and diet on the mouse gut microbiome and metabolome. Nat Microbiol 2016; 2:16221. [PMID: 27892936 DOI: 10.1038/nmicrobiol.2016.221] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/07/2016] [Indexed: 12/22/2022]
Abstract
Although the gut microbiome plays important roles in host physiology, health and disease1, we lack understanding of the complex interplay between host genetics and early life environment on the microbial and metabolic composition of the gut. We used the genetically diverse Collaborative Cross mouse system2 to discover that early life history impacts the microbiome composition, whereas dietary changes have only a moderate effect. By contrast, the gut metabolome was shaped mostly by diet, with specific non-dietary metabolites explained by microbial metabolism. Quantitative trait analysis identified mouse genetic trait loci (QTL) that impact the abundances of specific microbes. Human orthologues of genes in the mouse QTL are implicated in gastrointestinal cancer. Additionally, genes located in mouse QTL for Lactobacillales abundance are implicated in arthritis, rheumatic disease and diabetes. Furthermore, Lactobacillales abundance was predictive of higher host T-helper cell counts, suggesting an important link between Lactobacillales and host adaptive immunity.
Collapse
Affiliation(s)
- Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Sasha A Langley
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Young-Mo Kim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Colin J Brislawn
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Cecilia Noecker
- Department of Genome Sciences, University of Washington, Seattle, Washington 98105, USA
| | - Erika M Zink
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Sarah J Fansler
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Cameron P Casey
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Darla R Miller
- Systems Genetics Core Facility, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yurong Huang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Gary H Karpen
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | - Susan E Celniker
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - James B Brown
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Elhanan Borenstein
- Department of Genome Sciences, University of Washington, Seattle, Washington 98105, USA
- Department of Computer Science and Engineering, University of Washington, Seattle, Washington 98195, USA
- Santa Fe Institute, Santa Fe, New Mexico 87501, USA
| | - Janet K Jansson
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Thomas O Metz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| |
Collapse
|
409
|
Javierre BM, Burren OS, Wilder SP, Kreuzhuber R, Hill SM, Sewitz S, Cairns J, Wingett SW, Várnai C, Thiecke MJ, Burden F, Farrow S, Cutler AJ, Rehnström K, Downes K, Grassi L, Kostadima M, Freire-Pritchett P, Wang F, Stunnenberg HG, Todd JA, Zerbino DR, Stegle O, Ouwehand WH, Frontini M, Wallace C, Spivakov M, Fraser P. Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters. Cell 2016; 167:1369-1384.e19. [PMID: 27863249 PMCID: PMC5123897 DOI: 10.1016/j.cell.2016.09.037] [Citation(s) in RCA: 693] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/06/2016] [Accepted: 09/22/2016] [Indexed: 12/20/2022]
Abstract
Long-range interactions between regulatory elements and gene promoters play key roles in transcriptional regulation. The vast majority of interactions are uncharted, constituting a major missing link in understanding genome control. Here, we use promoter capture Hi-C to identify interacting regions of 31,253 promoters in 17 human primary hematopoietic cell types. We show that promoter interactions are highly cell type specific and enriched for links between active promoters and epigenetically marked enhancers. Promoter interactomes reflect lineage relationships of the hematopoietic tree, consistent with dynamic remodeling of nuclear architecture during differentiation. Interacting regions are enriched in genetic variants linked with altered expression of genes they contact, highlighting their functional role. We exploit this rich resource to connect non-coding disease variants to putative target promoters, prioritizing thousands of disease-candidate genes and implicating disease pathways. Our results demonstrate the power of primary cell promoter interactomes to reveal insights into genomic regulatory mechanisms underlying common diseases.
Collapse
Affiliation(s)
- Biola M Javierre
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Oliver S Burren
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Steven P Wilder
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Roman Kreuzhuber
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Steven M Hill
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Sven Sewitz
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Jonathan Cairns
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Steven W Wingett
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Csilla Várnai
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Michiel J Thiecke
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Frances Burden
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Antony J Cutler
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Karola Rehnström
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Myrto Kostadima
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK
| | - Paula Freire-Pritchett
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Fan Wang
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Geert Grooteplein Zuid 30, 6525 GA Nijmegen, the Netherlands
| | - John A Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Daniel R Zerbino
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK; Department of Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1HH, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge CB2 0PT, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Chris Wallace
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK; MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK; Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0SP, UK.
| | - Mikhail Spivakov
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
410
|
Genetic Predisposition to an Impaired Metabolism of the Branched-Chain Amino Acids and Risk of Type 2 Diabetes: A Mendelian Randomisation Analysis. PLoS Med 2016; 13:e1002179. [PMID: 27898682 PMCID: PMC5127513 DOI: 10.1371/journal.pmed.1002179] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/20/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Higher circulating levels of the branched-chain amino acids (BCAAs; i.e., isoleucine, leucine, and valine) are strongly associated with higher type 2 diabetes risk, but it is not known whether this association is causal. We undertook large-scale human genetic analyses to address this question. METHODS AND FINDINGS Genome-wide studies of BCAA levels in 16,596 individuals revealed five genomic regions associated at genome-wide levels of significance (p < 5 × 10-8). The strongest signal was 21 kb upstream of the PPM1K gene (beta in standard deviations [SDs] of leucine per allele = 0.08, p = 3.9 × 10-25), encoding an activator of the mitochondrial branched-chain alpha-ketoacid dehydrogenase (BCKD) responsible for the rate-limiting step in BCAA catabolism. In another analysis, in up to 47,877 cases of type 2 diabetes and 267,694 controls, a genetically predicted difference of 1 SD in amino acid level was associated with an odds ratio for type 2 diabetes of 1.44 (95% CI 1.26-1.65, p = 9.5 × 10-8) for isoleucine, 1.85 (95% CI 1.41-2.42, p = 7.3 × 10-6) for leucine, and 1.54 (95% CI 1.28-1.84, p = 4.2 × 10-6) for valine. Estimates were highly consistent with those from prospective observational studies of the association between BCAA levels and incident type 2 diabetes in a meta-analysis of 1,992 cases and 4,319 non-cases. Metabolome-wide association analyses of BCAA-raising alleles revealed high specificity to the BCAA pathway and an accumulation of metabolites upstream of branched-chain alpha-ketoacid oxidation, consistent with reduced BCKD activity. Limitations of this study are that, while the association of genetic variants appeared highly specific, the possibility of pleiotropic associations cannot be entirely excluded. Similar to other complex phenotypes, genetic scores used in the study captured a limited proportion of the heritability in BCAA levels. Therefore, it is possible that only some of the mechanisms that increase BCAA levels or affect BCAA metabolism are implicated in type 2 diabetes. CONCLUSIONS Evidence from this large-scale human genetic and metabolomic study is consistent with a causal role of BCAA metabolism in the aetiology of type 2 diabetes.
Collapse
|
411
|
Goswami S, Yee SW, Xu F, Sridhar SB, Mosley JD, Takahashi A, Kubo M, Maeda S, Davis RL, Roden DM, Hedderson MM, Giacomini KM, Savic RM. A Longitudinal HbA1c Model Elucidates Genes Linked to Disease Progression on Metformin. Clin Pharmacol Ther 2016; 100:537-547. [PMID: 27415606 PMCID: PMC5534241 DOI: 10.1002/cpt.428] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022]
Abstract
One-third of type-2 diabetic patients respond poorly to metformin. Despite extensive research, the impact of genetic and nongenetic factors on long-term outcome is unknown. In this study we combine nonlinear mixed effect modeling with computational genetic methodologies to identify predictors of long-term response. In all, 1,056 patients contributed their genetic, demographic, and long-term HbA1c data. The top nine variants (of 12,000 variants in 267 candidate genes) accounted for approximately one-third of the variability in the disease progression parameter. Average serum creatinine level, age, and weight were determinants of symptomatic response; however, explaining negligible variability. Two single nucleotide polymorphisms (SNPs) in CSMD1 gene (rs2617102, rs2954625) and one SNP in a pharmacologically relevant SLC22A2 gene (rs316009) influenced disease progression, with minor alleles leading to less and more favorable outcomes, respectively. Overall, our study highlights the influence of genetic factors on long-term HbA1c response and provides a computational model, which when validated, may be used to individualize treatment.
Collapse
Affiliation(s)
- S Goswami
- University of California, San Francisco, San Francisco, California, USA
| | - S W Yee
- University of California, San Francisco, San Francisco, California, USA
| | - F Xu
- Kaiser Permanente Northern California, Oakland, California, USA
| | - S B Sridhar
- Kaiser Permanente Northern California, Oakland, California, USA
| | - J D Mosley
- Vanderbilt University, Nashville, Tennessee, USA
| | - A Takahashi
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - M Kubo
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - S Maeda
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - R L Davis
- Kaiser Permanente Georgia, Atlanta, Georgia, USA
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - D M Roden
- Vanderbilt University, Nashville, Tennessee, USA
| | - M M Hedderson
- Kaiser Permanente Northern California, Oakland, California, USA
| | - K M Giacomini
- University of California, San Francisco, San Francisco, California, USA.
| | - R M Savic
- University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
412
|
Pedersen HK, Gudmundsdottir V, Pedersen MK, Brorsson C, Brunak S, Gupta R. Ranking factors involved in diabetes remission after bariatric surgery using machine-learning integrating clinical and genomic biomarkers. NPJ Genom Med 2016; 1:16035. [PMID: 29263820 PMCID: PMC5685313 DOI: 10.1038/npjgenmed.2016.35] [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: 11/29/2015] [Revised: 08/22/2016] [Accepted: 08/25/2016] [Indexed: 01/07/2023] Open
Abstract
As weight-loss surgery is an effective treatment for the glycaemic control of type 2 diabetes in obese patients, yet not all patients benefit, it is valuable to find predictive factors for this diabetic remission. This will help elucidating possible mechanistic insights and form the basis for prioritising obese patients with dysregulated diabetes for surgery where diabetes remission is of interest. In this study, we combine both clinical and genomic factors using heuristic methods, informed by prior biological knowledge in order to rank factors that would have a role in predicting diabetes remission, and indeed in identifying patients who may have low likelihood in responding to bariatric surgery for improved glycaemic control. Genetic variants from the Illumina CardioMetaboChip were prioritised through single-association tests and then seeded a larger selection from protein-protein interaction networks. Artificial neural networks allowing nonlinear correlations were trained to discriminate patients with and without surgery-induced diabetes remission, and the importance of each clinical and genetic parameter was evaluated. The approach highlighted insulin treatment, baseline HbA1c levels, use of insulin-sensitising agents and baseline serum insulin levels, as the most informative variables with a decent internal validation performance (74% accuracy, area under the curve (AUC) 0.81). Adding information for the eight top-ranked single nucleotide polymorphisms (SNPs) significantly boosted classification performance to 84% accuracy (AUC 0.92). The eight SNPs mapped to eight genes - ABCA1, ARHGEF12, CTNNBL1, GLI3, PROK2, RYBP, SMUG1 and STXBP5 - three of which are known to have a role in insulin secretion, insulin sensitivity or obesity, but have not been indicated for diabetes remission after bariatric surgery before.
Collapse
Affiliation(s)
- Helle Krogh Pedersen
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Valborg Gudmundsdottir
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mette Krogh Pedersen
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Brorsson
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Søren Brunak
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ramneek Gupta
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
413
|
Langlois C, Abadi A, Peralta-Romero J, Alyass A, Suarez F, Gomez-Zamudio J, Burguete-Garcia AI, Yazdi FT, Cruz M, Meyre D. Evaluating the transferability of 15 European-derived fasting plasma glucose SNPs in Mexican children and adolescents. Sci Rep 2016; 6:36202. [PMID: 27782183 PMCID: PMC5080582 DOI: 10.1038/srep36202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/12/2016] [Indexed: 12/15/2022] Open
Abstract
Genome wide association studies (GWAS) have identified single-nucleotide polymorphisms (SNPs) that are associated with fasting plasma glucose (FPG) in adult European populations. The contribution of these SNPs to FPG in non-Europeans and children is unclear. We studied the association of 15 GWAS SNPs and a genotype score (GS) with FPG and 7 metabolic traits in 1,421 Mexican children and adolescents from Mexico City. Genotyping of the 15 SNPs was performed using TaqMan Open Array. We used multivariate linear regression models adjusted for age, sex, body mass index standard deviation score, and recruitment center. We identified significant associations between 3 SNPs (G6PC2 (rs560887), GCKR (rs1260326), MTNR1B (rs10830963)), the GS and FPG level. The FPG risk alleles of 11 out of the 15 SNPs (73.3%) displayed significant or non-significant beta values for FPG directionally consistent with those reported in adult European GWAS. The risk allele frequencies for 11 of 15 (73.3%) SNPs differed significantly in Mexican children and adolescents compared to European adults from the 1000G Project, but no significant enrichment in FPG risk alleles was observed in the Mexican population. Our data support a partial transferability of European GWAS FPG association signals in children and adolescents from the admixed Mexican population.
Collapse
Affiliation(s)
- Christine Langlois
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Arkan Abadi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Jesus Peralta-Romero
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Akram Alyass
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Fernando Suarez
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Jaime Gomez-Zamudio
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ana I. Burguete-Garcia
- Centro de investigación sobre enfermedades infecciosas. Instituto Nacional de Salud Pública. Cuernavaca, Morelos, Mexico
| | - Fereshteh T. Yazdi
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Miguel Cruz
- Medical Research Unit in Biochemistry, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
414
|
Lotta LA, Sharp SJ, Burgess S, Perry JRB, Stewart ID, Willems SM, Luan J, Ardanaz E, Arriola L, Balkau B, Boeing H, Deloukas P, Forouhi NG, Franks PW, Grioni S, Kaaks R, Key TJ, Navarro C, Nilsson PM, Overvad K, Palli D, Panico S, Quirós JR, Riboli E, Rolandsson O, Sacerdote C, Salamanca EC, Slimani N, Spijkerman AMW, Tjonneland A, Tumino R, van der A DL, van der Schouw YT, McCarthy MI, Barroso I, O’Rahilly S, Savage DB, Sattar N, Langenberg C, Scott RA, Wareham NJ. Association Between Low-Density Lipoprotein Cholesterol-Lowering Genetic Variants and Risk of Type 2 Diabetes: A Meta-analysis. JAMA 2016; 316:1383-1391. [PMID: 27701660 PMCID: PMC5386134 DOI: 10.1001/jama.2016.14568] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Low-density lipoprotein cholesterol (LDL-C)-lowering alleles in or near NPC1L1 or HMGCR, encoding the respective molecular targets of ezetimibe and statins, have previously been used as proxies to study the efficacy of these lipid-lowering drugs. Alleles near HMGCR are associated with a higher risk of type 2 diabetes, similar to the increased incidence of new-onset diabetes associated with statin treatment in randomized clinical trials. It is unknown whether alleles near NPC1L1 are associated with the risk of type 2 diabetes. Objective To investigate whether LDL-C-lowering alleles in or near NPC1L1 and other genes encoding current or prospective molecular targets of lipid-lowering therapy (ie, HMGCR, PCSK9, ABCG5/G8, LDLR) are associated with the risk of type 2 diabetes. Design, Setting, and Participants The associations with type 2 diabetes and coronary artery disease of LDL-C-lowering genetic variants were investigated in meta-analyses of genetic association studies. Meta-analyses included 50 775 individuals with type 2 diabetes and 270 269 controls and 60 801 individuals with coronary artery disease and 123 504 controls. Data collection took place in Europe and the United States between 1991 and 2016. Exposures Low-density lipoprotein cholesterol-lowering alleles in or near NPC1L1, HMGCR, PCSK9, ABCG5/G8, and LDLR. Main Outcomes and Measures Odds ratios (ORs) for type 2 diabetes and coronary artery disease. Results Low-density lipoprotein cholesterol-lowering genetic variants at NPC1L1 were inversely associated with coronary artery disease (OR for a genetically predicted 1-mmol/L [38.7-mg/dL] reduction in LDL-C of 0.61 [95% CI, 0.42-0.88]; P = .008) and directly associated with type 2 diabetes (OR for a genetically predicted 1-mmol/L reduction in LDL-C of 2.42 [95% CI, 1.70-3.43]; P < .001). For PCSK9 genetic variants, the OR for type 2 diabetes per 1-mmol/L genetically predicted reduction in LDL-C was 1.19 (95% CI, 1.02-1.38; P = .03). For a given reduction in LDL-C, genetic variants were associated with a similar reduction in coronary artery disease risk (I2 = 0% for heterogeneity in genetic associations; P = .93). However, associations with type 2 diabetes were heterogeneous (I2 = 77.2%; P = .002), indicating gene-specific associations with metabolic risk of LDL-C-lowering alleles. Conclusions and Relevance In this meta-analysis, exposure to LDL-C-lowering genetic variants in or near NPC1L1 and other genes was associated with a higher risk of type 2 diabetes. These data provide insights into potential adverse effects of LDL-C-lowering therapy.
Collapse
Affiliation(s)
- Luca A. Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Stephen. J Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Isobel. D Stewart
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Sara M. Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Jian’an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Eva Ardanaz
- Navarre Public Health Institute (ISPN), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA) Pamplona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Larraitz Arriola
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Public Health Division of Gipuzkoa, San Sebastian, Spain
- Instituto BIO-Donostia, Basque Government, San Sebastian, Spain
| | | | - Heiner Boeing
- German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Panos Deloukas
- The Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Paul W Franks
- Lund University, Malmö, Sweden
- Umeå University, Umeå, Sweden
| | - Sara Grioni
- Epidemiology and Prevention Unit, Milan, Italy
| | - Rudolf Kaaks
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | | | - Carmen Navarro
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Unit of Preventive Medicine and Public Health, School of Medicine, University of Murcia, Spain
| | | | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
- Aalborg University Hospital, Aalborg, Denmark
| | - Domenico Palli
- Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | | | - Elio Riboli
- School of Public Health, Imperial College London, United Kingdom
| | | | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Citta' della Salute e della Scienza Hospital-University of Turin and Center for Cancer Prevention (CPO), Torino, Italy
- Human Genetics Foundation (HuGeF), Torino, Italy
| | - Elena C Salamanca
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Andalusian School of Public Health, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Granada.ibs), Granada, Spain
| | - Nadia Slimani
- International Agency for Research on Cancer, Lyon, France
| | | | | | | | - Daphne L van der A
- National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | | | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Inês Barroso
- The Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Stephen O’Rahilly
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - David. B Savage
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, United Kingdom
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Robert. A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
415
|
Marinelli M, Pappa I, Bustamante M, Bonilla C, Suarez A, Tiesler CM, Vilor-Tejedor N, Zafarmand MH, Alvarez-Pedrerol M, Andersson S, Bakermans-Kranenburg MJ, Estivill X, Evans DM, Flexeder C, Forns J, Gonzalez JR, Guxens M, Huss A, van IJzendoorn MH, Jaddoe VW, Julvez J, Lahti J, López-Vicente M, Lopez-Espinosa MJ, Manz J, Mileva-Seitz VR, Perola M, Pesonen AK, Rivadeneira F, Salo PP, Shahand S, Schulz H, Standl M, Thiering E, Timpson NJ, Torrent M, Uitterlinden AG, Smith GD, Estarlich M, Heinrich J, Räikkönen K, Vrijkotte TG, Tiemeier H, Sunyer J. Heritability and Genome-Wide Association Analyses of Sleep Duration in Children: The EAGLE Consortium. Sleep 2016; 39:1859-1869. [PMID: 27568811 PMCID: PMC5020368 DOI: 10.5665/sleep.6170] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 06/09/2016] [Indexed: 01/20/2023] Open
Abstract
STUDY OBJECTIVES Low or excessive sleep duration has been associated with multiple outcomes, but the biology behind these associations remains elusive. Specifically, genetic studies in children are scarce. In this study, we aimed to: (1) estimate the proportion of genetic variance of sleep duration in children attributed to common single nucleotide polymorphisms (SNPs), (2) identify novel SNPs associated with sleep duration in children, and (3) investigate the genetic overlap of sleep duration in children and related metabolic and psychiatric traits. METHODS We performed a population-based molecular genetic study, using data form the EArly Genetics and Life course Epidemiology (EAGLE) Consortium. 10,554 children of European ancestry were included in the discovery, and 1,250 children in the replication phase. RESULTS We found evidence of significant but modest SNP heritability of sleep duration in children (SNP h2 0.14, 95% CI [0.05, 0.23]) using the LD score regression method. A novel region at chromosome 11q13.4 (top SNP: rs74506765, P = 2.27e-08) was associated with sleep duration in children, but this was not replicated in independent studies. Nominally significant genetic overlap was only found (rG = 0.23, P = 0.05) between sleep duration in children and type 2 diabetes in adults, supporting the hypothesis of a common pathogenic mechanism. CONCLUSIONS The significant SNP heritability of sleep duration in children and the suggestive genetic overlap with type 2 diabetes support the search for genetic mechanisms linking sleep duration in children to multiple outcomes in health and disease.
Collapse
Affiliation(s)
- Marcella Marinelli
- Agency for Healthcare Quality and Evaluation of Catalonia (AQuAS), Roc Boronat, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
| | - Irene Pappa
- School of Pedagogical and Educational Sciences, Erasmus University Rotterdam, The Netherlands
- Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Mariona Bustamante
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Centre for Genomic Regulation (CRG), the Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carolina Bonilla
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Anna Suarez
- Institute of behavioural sciences, University of Helsinki, Helsinki, Finland
| | - Carla M. Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Natalia Vilor-Tejedor
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Mohammad Hadi Zafarmand
- Department of Public Health, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Academic Medical Centre, University of Amsterdam, The Netherlands
| | - Mar Alvarez-Pedrerol
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sture Andersson
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | | | - Xavier Estivill
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centre for Genomic Regulation (CRG), the Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut Hospital del Mar d'Investigacions Mediques (IMIM), 08003 Barcelona, Spain
| | - David M. Evans
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Claudia Flexeder
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Joan Forns
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Juan R. Gonzalez
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Monica Guxens
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Anke Huss
- Institute for Risk Assessment Sciences, Utrecht University, the Netherlands
| | - Marinus H. van IJzendoorn
- School of Pedagogical and Educational Sciences, Erasmus University Rotterdam, The Netherlands
- Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Centre for Child and Family Studies, Leiden University, Leiden, The Netherlands
| | - Vincent W.V. Jaddoe
- Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus Medical Center- Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jordi Julvez
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jari Lahti
- Institute of behavioural sciences, University of Helsinki, Helsinki, Finland
- Helsinki Collegium for Advanced Studies, Helsinki, Finland
- Folkhälsan Research Centre, Finland
| | - Mónica López-Vicente
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Maria-Jose Lopez-Espinosa
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO, Universitat Jaume I, Universitat de València, Spain
| | - Judith Manz
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Markus Perola
- Public Health Genomics Unit and Institute for Molecular Medicine FIMM, University of Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | | | - Fernando Rivadeneira
- Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Perttu P. Salo
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Shayan Shahand
- Department of Clinical Epidemiology Biostatistics and Bioinformatics, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Nicholas J. Timpson
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | | | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - George Davey Smith
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Marisa Estarlich
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO, Universitat Jaume I, Universitat de València, Spain
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Katri Räikkönen
- Institute of behavioural sciences, University of Helsinki, Helsinki, Finland
| | - Tanja G.M. Vrijkotte
- Department of Public Health, Academic Medical Center (AMC), University of Amsterdam, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jordi Sunyer
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut Hospital del Mar d'Investigacions Mediques (IMIM), 08003 Barcelona, Spain
- Address correspondence to: Jordi Sunyer, PhD,
ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Doctor Aiguader, 88, E-08003 Barcelona, Spain+34 93 214 73 00+ 34 93 214 73 02
| |
Collapse
|
416
|
Walford GA, Gustafsson S, Rybin D, Stančáková A, Chen H, Liu CT, Hong J, Jensen RA, Rice K, Morris AP, Mägi R, Tönjes A, Prokopenko I, Kleber ME, Delgado G, Silbernagel G, Jackson AU, Appel EV, Grarup N, Lewis JP, Montasser ME, Landenvall C, Staiger H, Luan J, Frayling TM, Weedon MN, Xie W, Morcillo S, Martínez-Larrad MT, Biggs ML, Chen YDI, Corbaton-Anchuelo A, Færch K, Gómez-Zumaquero JM, Goodarzi MO, Kizer JR, Koistinen HA, Leong A, Lind L, Lindgren C, Machicao F, Manning AK, Martín-Núñez GM, Rojo-Martínez G, Rotter JI, Siscovick DS, Zmuda JM, Zhang Z, Serrano-Rios M, Smith U, Soriguer F, Hansen T, Jørgensen TJ, Linnenberg A, Pedersen O, Walker M, Langenberg C, Scott RA, Wareham NJ, Fritsche A, Häring HU, Stefan N, Groop L, O'Connell JR, Boehnke M, Bergman RN, Collins FS, Mohlke KL, Tuomilehto J, März W, Kovacs P, Stumvoll M, Psaty BM, Kuusisto J, Laakso M, Meigs JB, Dupuis J, Ingelsson E, Florez JC. Genome-Wide Association Study of the Modified Stumvoll Insulin Sensitivity Index Identifies BCL2 and FAM19A2 as Novel Insulin Sensitivity Loci. Diabetes 2016; 65:3200-11. [PMID: 27416945 PMCID: PMC5033262 DOI: 10.2337/db16-0199] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/05/2016] [Indexed: 01/19/2023]
Abstract
Genome-wide association studies (GWAS) have found few common variants that influence fasting measures of insulin sensitivity. We hypothesized that a GWAS of an integrated assessment of fasting and dynamic measures of insulin sensitivity would detect novel common variants. We performed a GWAS of the modified Stumvoll Insulin Sensitivity Index (ISI) within the Meta-Analyses of Glucose and Insulin-Related Traits Consortium. Discovery for genetic association was performed in 16,753 individuals, and replication was attempted for the 23 most significant novel loci in 13,354 independent individuals. Association with ISI was tested in models adjusted for age, sex, and BMI and in a model analyzing the combined influence of the genotype effect adjusted for BMI and the interaction effect between the genotype and BMI on ISI (model 3). In model 3, three variants reached genome-wide significance: rs13422522 (NYAP2; P = 8.87 × 10(-11)), rs12454712 (BCL2; P = 2.7 × 10(-8)), and rs10506418 (FAM19A2; P = 1.9 × 10(-8)). The association at NYAP2 was eliminated by conditioning on the known IRS1 insulin sensitivity locus; the BCL2 and FAM19A2 associations were independent of known cardiometabolic loci. In conclusion, we identified two novel loci and replicated known variants associated with insulin sensitivity. Further studies are needed to clarify the causal variant and function at the BCL2 and FAM19A2 loci.
Collapse
Affiliation(s)
- Geoffrey A Walford
- Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston, MA Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA Department of Medicine, Harvard Medical School, Boston, MA
| | | | - Denis Rybin
- Data Coordinating Center, Boston University School of Public Health, Boston, MA
| | - Alena Stančáková
- University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Han Chen
- Department of Biostatistics, Boston University School of Public Health, Boston, MA Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Richard A Jensen
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA Department of Medicine, University of Washington, Seattle, WA
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, U.K. Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K. Department of Genomics of Common Disease, Imperial College London, London, U.K. Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
| | - Marcus E Kleber
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Graciela Delgado
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| | - Emil V Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joshua P Lewis
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - May E Montasser
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Claes Landenvall
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Harald Staiger
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany German Center for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | | | | | - Weijia Xie
- University of Exeter Medical School, Exeter, U.K
| | - Sonsoles Morcillo
- CIBER Pathophysiology of Obesity and Nutrition, Madrid, Spain Department of Endocrinology and Nutrition, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - María Teresa Martínez-Larrad
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Mary L Biggs
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA Department of Biostatistics, University of Washington, Seattle, WA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA
| | - Arturo Corbaton-Anchuelo
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | | | - Juan Miguel Gómez-Zumaquero
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain Sequencing and Genotyping Platform, Hospital Carlos Haya de Málaga, Málaga, Spain
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Heikki A Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Aaron Leong
- Department of Medicine, Harvard Medical School, Boston, MA Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K. Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Fausto Machicao
- German Center for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Alisa K Manning
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA Department of Medicine, Harvard Medical School, Boston, MA Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Gracia María Martín-Núñez
- Department of Endocrinology and Nutrition, Hospitales Regional Universitario y Virgen de la Victoria de Málaga, Málaga, Spain
| | - Gemma Rojo-Martínez
- Department of Endocrinology and Nutrition, Hospital Regional Universitario de Málaga, Málaga, Spain Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA
| | - David S Siscovick
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA Department of Medicine, University of Washington, Seattle, WA Department of Epidemiology, University of Washington, Seattle, WA The New York Academy of Medicine, New York, NY
| | - Joseph M Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Zhongyang Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Manuel Serrano-Rios
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Federico Soriguer
- Department of Endocrinology and Nutrition, Hospital Regional Universitario de Málaga, Málaga, Spain Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben J Jørgensen
- Department of Public Health, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark Faculty of Medicine, Aalborg University, Aalborg, Denmark Research Center for Prevention and Health, The Capital Region of Denmark, Copenhagen, Denmark
| | - Allan Linnenberg
- Research Center for Prevention and Health, The Capital Region of Denmark, Copenhagen, Denmark Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany German Center for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany German Center for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany German Center for Diabetes Research (DZD), Tübingen, Germany Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Jeff R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| | - Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Jaakko Tuomilehto
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland Centre for Vascular Prevention, Danube-University Krems, Krems, Austria Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia Dasman Diabetes Institute, Dasman, Kuwait
| | - Winfried März
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria Synlab Academy, Synlab Services GmbH, Mannheim and Augsburg, Germany
| | - Peter Kovacs
- Integrated Research and Treatment (IFB) Center AdiposityDiseases, University of Leipzig, Leipzig, Germany
| | | | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA Department of Medicine, University of Washington, Seattle, WA Epidemiology and Health Services, University of Washington, Seattle, WA Group Health Research Institute, Seattle, WA Group Health Cooperation, Seattle, WA
| | - 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
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jose C Florez
- Diabetes Research Center (Diabetes Unit), Massachusetts General Hospital, Boston, MA Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
417
|
Corbin LJ, Richmond RC, Wade KH, Burgess S, Bowden J, Smith GD, Timpson NJ. BMI as a Modifiable Risk Factor for Type 2 Diabetes: Refining and Understanding Causal Estimates Using Mendelian Randomization. Diabetes 2016; 65:3002-7. [PMID: 27402723 PMCID: PMC5279886 DOI: 10.2337/db16-0418] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/05/2016] [Indexed: 12/20/2022]
Abstract
This study focused on resolving the relationship between BMI and type 2 diabetes. The availability of multiple variants associated with BMI offers a new chance to resolve the true causal effect of BMI on type 2 diabetes; however, the properties of these associations and their validity as genetic instruments need to be considered alongside established and new methods for undertaking Mendelian randomization (MR). We explore the potential for pleiotropic genetic variants to generate bias, revise existing estimates, and illustrate value in new analysis methods. A two-sample MR approach with 96 genetic variants was used with three different analysis methods, two of which (MR-Egger and the weighted median) have been developed specifically to address problems of invalid instrumental variables. We estimate an odds ratio for type 2 diabetes per unit increase in BMI (kg/m(2)) of between 1.19 and 1.38, with the most stable estimate using all instruments and a weighted median approach (1.26 [95% CI 1.17, 1.34]). TCF7L2(rs7903146) was identified as a complex effect or pleiotropic instrument, and removal of this variant resulted in convergence of causal effect estimates from different causal analysis methods. This indicated the potential for pleiotropy to affect estimates and differences in performance of alternative analytical methods. In a real type 2 diabetes-focused example, this study demonstrates the potential impact of invalid instruments on causal effect estimates and the potential for new approaches to mitigate the bias caused.
Collapse
Affiliation(s)
- Laura J Corbin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | | | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - Stephen Burgess
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K. Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Jack Bowden
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K. MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, U.K
| | | | | |
Collapse
|
418
|
Abstract
Despite the progress made in understanding the biology of autism spectrum disorder (ASD), effective biological interventions for the core symptoms remain elusive. Because of the etiological heterogeneity of ASD, identification of a "one-size-fits-all" treatment approach will likely continue to be challenging. A meeting was convened at the University of Missouri and the Thompson Center to discuss strategies for stratifying patients with ASD for the purpose of moving toward precision medicine. The "white paper" presented here articulates the challenges involved and provides suggestions for future solutions.
Collapse
|
419
|
Sandholm N, Van Zuydam N, Ahlqvist E, Juliusdottir T, Deshmukh HA, Rayner NW, Di Camillo B, Forsblom C, Fadista J, Ziemek D, Salem RM, Hiraki LT, Pezzolesi M, Trégouët D, Dahlström E, Valo E, Oskolkov N, Ladenvall C, Marcovecchio ML, Cooper J, Sambo F, Malovini A, Manfrini M, McKnight AJ, Lajer M, Harjutsalo V, Gordin D, Parkkonen M, Tuomilehto J, Lyssenko V, McKeigue PM, Rich SS, Brosnan MJ, Fauman E, Bellazzi R, Rossing P, Hadjadj S, Krolewski A, Paterson AD, Florez JC, Hirschhorn JN, Maxwell AP, Dunger D, Cobelli C, Colhoun HM, Groop L, McCarthy MI, Groop PH. The Genetic Landscape of Renal Complications in Type 1 Diabetes. J Am Soc Nephrol 2016; 28:557-574. [PMID: 27647854 DOI: 10.1681/asn.2016020231] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022] Open
Abstract
Diabetes is the leading cause of ESRD. Despite evidence for a substantial heritability of diabetic kidney disease, efforts to identify genetic susceptibility variants have had limited success. We extended previous efforts in three dimensions, examining a more comprehensive set of genetic variants in larger numbers of subjects with type 1 diabetes characterized for a wider range of cross-sectional diabetic kidney disease phenotypes. In 2843 subjects, we estimated that the heritability of diabetic kidney disease was 35% (P=6.4×10-3). Genome-wide association analysis and replication in 12,540 individuals identified no single variants reaching stringent levels of significance and, despite excellent power, provided little independent confirmation of previously published associated variants. Whole-exome sequencing in 997 subjects failed to identify any large-effect coding alleles of lower frequency influencing the risk of diabetic kidney disease. However, sets of alleles increasing body mass index (P=2.2×10-5) and the risk of type 2 diabetes (P=6.1×10-4) associated with the risk of diabetic kidney disease. We also found genome-wide genetic correlation between diabetic kidney disease and failure at smoking cessation (P=1.1×10-4). Pathway analysis implicated ascorbate and aldarate metabolism (P=9.0×10-6), and pentose and glucuronate interconversions (P=3.0×10-6) in pathogenesis of diabetic kidney disease. These data provide further evidence for the role of genetic factors influencing diabetic kidney disease in those with type 1 diabetes and highlight some key pathways that may be responsible. Altogether these results reveal important biology behind the major cause of kidney disease.
Collapse
Affiliation(s)
- Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Natalie Van Zuydam
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Medical Research Institute
| | - Emma Ahlqvist
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Harshal A Deshmukh
- Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - N William Rayner
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Barbara Di Camillo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Joao Fadista
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Daniel Ziemek
- Computational Sciences, Pfizer Worldwide Research and Development, Berlin, Germany
| | - Rany M Salem
- Departments of Genetics,Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Divisions of Endocrinology and Genetics, Boston Children's Hospital, Boston, Massachusetts
| | - Linda T Hiraki
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marcus Pezzolesi
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, Massachusetts
| | - David Trégouët
- Sorbonne Universities, Pierre et Marie Curie University (UPMC) and National Institute for Health and Medical Research, Mixed Research Unit in Health (UMR_S) 1166, Paris, France.,Institute for Cardiometabolism and Nutrition, Genomics and pathophysiology of Cardiovascular diseases, Paris, France
| | - Emma Dahlström
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Erkka Valo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Nikolay Oskolkov
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Claes Ladenvall
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Jason Cooper
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Francesco Sambo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alberto Malovini
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.,Laboratory of Informatics and Systems Engineering for Clinical Research, Scientific Institute for Research, Hospitalization and Health Care, IRCCS (Instituto di Ricovero e Cura a Carattere Scientifico); Salvatore Maugeri Foundation, Pavia, Italy
| | - Marco Manfrini
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Amy Jayne McKnight
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, United Kingdom
| | - Maria Lajer
- Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Daniel Gordin
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Maija Parkkonen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | | | - Jaakko Tuomilehto
- The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland.,Centre for Vascular Prevention, Danube University Krems, Krems, Austria
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden.,Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark
| | - Paul M McKeigue
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | | | - Eric Fauman
- Computational Sciences, Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Riccardo Bellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Peter Rossing
- Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark.,Department of Health, Aarhus University, Aarhus, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Samy Hadjadj
- Functional Research Unit of Medicine and Pharmacy, University of Poitiers, Poitiers, France.,Department of Endocrinology-Diabetology and Center of Clinical Investigation, Poitiers University Hospital, Poitiers, France.,Institute National pour la Santé et la Recherche Médicale, National Institute for Health and Medical Research, Center of Clinical Investigation 1402 and Unit 1082, Poitiers, France
| | - Andrzej Krolewski
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, Massachusetts
| | - Andrew D Paterson
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
| | - Joel N Hirschhorn
- Departments of Genetics,Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Divisions of Endocrinology and Genetics, Boston Children's Hospital, Boston, Massachusetts
| | - Alexander P Maxwell
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, United Kingdom.,Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom; and
| | | | - David Dunger
- Department of Paediatrics, Institute of Metabolic Science, and
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Helen M Colhoun
- Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Baker IDI (International Diabetes Institute) Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | |
Collapse
|
420
|
Functional Analysis of Mouse G6pc1 Mutations Using a Novel In Situ Assay for Glucose-6-Phosphatase Activity and the Effect of Mutations in Conserved Human G6PC1/G6PC2 Amino Acids on G6PC2 Protein Expression. PLoS One 2016; 11:e0162439. [PMID: 27611587 PMCID: PMC5017610 DOI: 10.1371/journal.pone.0162439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 08/23/2016] [Indexed: 11/19/2022] Open
Abstract
Elevated fasting blood glucose (FBG) has been associated with increased risk for development of type 2 diabetes. Single nucleotide polymorphisms (SNPs) in G6PC2 are the most important common determinants of variations in FBG in humans. Studies using G6pc2 knockout mice suggest that G6pc2 regulates the glucose sensitivity of insulin secretion. G6PC2 and the related G6PC1 and G6PC3 genes encode glucose-6-phosphatase catalytic subunits. This study describes a functional analysis of 22 non-synonymous G6PC2 SNPs, that alter amino acids that are conserved in human G6PC1, mouse G6pc1 and mouse G6pc2, with the goal of identifying variants that potentially affect G6PC2 activity/expression. Published data suggest strong conservation of catalytically important amino acids between all four proteins and the related G6PC3 isoform. Because human G6PC2 has very low glucose-6-phosphatase activity we used an indirect approach, examining the effect of these SNPs on mouse G6pc1 activity. Using a novel in situ functional assay for glucose-6-phosphatase activity we demonstrate that the amino acid changes associated with the human G6PC2 rs144254880 (Arg79Gln), rs149663725 (Gly114Arg) and rs2232326 (Ser324Pro) SNPs reduce mouse G6pc1 enzyme activity without affecting protein expression. The Arg79Gln variant alters an amino acid mutation of which, in G6PC1, has previously been shown to cause glycogen storage disease type 1a. We also demonstrate that the rs368382511 (Gly8Glu), rs138726309 (His177Tyr), rs2232323 (Tyr207Ser) rs374055555 (Arg293Trp), rs2232326 (Ser324Pro), rs137857125 (Pro313Leu) and rs2232327 (Pro340Leu) SNPs confer decreased G6PC2 protein expression. In summary, these studies identify multiple G6PC2 variants that have the potential to be associated with altered FBG in humans.
Collapse
|
421
|
Glastonbury C, Viñuela A, Buil A, Halldorsson G, Thorleifsson G, Helgason H, Thorsteinsdottir U, Stefansson K, Dermitzakis E, Spector T, Small K. Adiposity-Dependent Regulatory Effects on Multi-tissue Transcriptomes. Am J Hum Genet 2016; 99:567-579. [PMID: 27588447 PMCID: PMC5011064 DOI: 10.1016/j.ajhg.2016.07.001] [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: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 10/25/2022] Open
Abstract
Obesity is a global epidemic that is causally associated with a range of diseases, including type 2 diabetes and cardiovascular disease, at the population-level. However, there is marked heterogeneity in obesity-related outcomes among individuals. This might reflect genotype-dependent responses to adiposity. Given that adiposity, measured by BMI, is associated with widespread changes in gene expression and regulatory variants mediate the majority of known complex trait loci, we sought to identify gene-by-BMI (G × BMI) interactions on the regulation of gene expression in a multi-tissue RNA-sequencing (RNA-seq) dataset from the TwinsUK cohort (n = 856). At a false discovery rate of 5%, we identified 16 cis G × BMI interactions (top cis interaction: CHURC1, rs7143432, p = 2.0 × 10(-12)) and one variant regulating 53 genes in trans (top trans interaction: ZNF423, rs3851570, p = 8.2 × 10(-13)), all in adipose tissue. The interactions were adipose-specific and enriched for variants overlapping adipocyte enhancers, and regulated genes were enriched for metabolic and inflammatory processes. We replicated a subset of the interactions in an independent adipose RNA-seq dataset (deCODE genetics, n = 754). We also confirmed the interactions with an alternate measure of obesity, dual-energy X-ray absorptiometry (DXA)-derived visceral-fat-volume measurements, in a subset of TwinsUK individuals (n = 682). The identified G × BMI regulatory effects demonstrate the dynamic nature of gene regulation and reveal a functional mechanism underlying the heterogeneous response to obesity. Additionally, we have provided a web browser allowing interactive exploration of the dataset, including of association between expression, BMI, and G × BMI regulatory effects in four tissues.
Collapse
|
422
|
Eze IC, Imboden M, Kumar A, von Eckardstein A, Stolz D, Gerbase MW, Künzli N, Pons M, Kronenberg F, Schindler C, Probst-Hensch N. Air pollution and diabetes association: Modification by type 2 diabetes genetic risk score. ENVIRONMENT INTERNATIONAL 2016; 94:263-271. [PMID: 27281273 DOI: 10.1016/j.envint.2016.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/11/2016] [Accepted: 04/22/2016] [Indexed: 05/26/2023]
Abstract
Exposure to ambient air pollution (AP) exposure has been linked to type 2 diabetes (T2D) risk. Evidence on the impact of T2D genetic variants on AP susceptibility is lacking. Compared to single variants, joint genetic variants contribute substantially to disease risk. We investigated the modification of AP and diabetes association by a genetic risk score (GRS) covering 63 T2D genes in 1524 first follow-up participants of the Swiss cohort study on air pollution and lung and heart diseases in adults. Genome-wide data and covariates were available from a nested asthma case-control study design. AP was estimated as 10-year mean residential particulate matter <10μm (PM10). We computed count-GRS and weighted-GRS, and applied PM10 interaction terms in mixed logistic regressions, on odds of diabetes. Analyses were stratified by pathways of diabetes pathology and by asthma status. Diabetes prevalence was 4.6% and mean exposure to PM10 was 22μg/m(3). Odds of diabetes increased by 8% (95% confidence interval: 2, 14%) per T2D risk allele and by 35% (-8, 97%) per 10μg/m(3) exposure to PM10. We observed a positive interaction between PM10 and count-GRS on diabetes [ORinteraction=1.10 (1.01, 1.20)], associations being strongest among participants at the highest quartile of count-GRS [OR: 1.97 (1.00, 3.87)]. Stronger interactions were observed with variants of the GRS involved in insulin resistance [(ORinteraction=1.22 (1.00, 1.50)] than with variants related to beta-cell function. Interactions with count-GRS were stronger among asthma cases. We observed similar results with weighted-GRS. Five single variants near GRB14, UBE2E2, PTPRD, VPS26A and KCNQ1 showed nominally significant interactions with PM10 (P<0.05). Our results suggest that genetic risk for T2D may modify susceptibility to air pollution through alterations in insulin sensitivity. These results need confirmation in diabetes cohort consortia.
Collapse
Affiliation(s)
- Ikenna C Eze
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Ashish Kumar
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Karolinska Institutet, Stockholm, Sweden
| | | | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland
| | | | - Nino Künzli
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Marco Pons
- Department of Internal Medicine, Regional Hospital of Lugano, Lugano, Switzerland
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Schindler
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
423
|
Franzén O, Ermel R, Cohain A, Akers NK, Di Narzo A, Talukdar HA, Foroughi-Asl H, Giambartolomei C, Fullard JF, Sukhavasi K, Köks S, Gan LM, Giannarelli C, Kovacic JC, Betsholtz C, Losic B, Michoel T, Hao K, Roussos P, Skogsberg J, Ruusalepp A, Schadt EE, Björkegren JLM. Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases. Science 2016; 353:827-30. [PMID: 27540175 DOI: 10.1126/science.aad6970] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 07/22/2016] [Indexed: 12/11/2022]
Abstract
Genome-wide association studies (GWAS) have identified hundreds of cardiometabolic disease (CMD) risk loci. However, they contribute little to genetic variance, and most downstream gene-regulatory mechanisms are unknown. We genotyped and RNA-sequenced vascular and metabolic tissues from 600 coronary artery disease patients in the Stockholm-Tartu Atherosclerosis Reverse Networks Engineering Task study (STARNET). Gene expression traits associated with CMD risk single-nucleotide polymorphism (SNPs) identified by GWAS were more extensively found in STARNET than in tissue- and disease-unspecific gene-tissue expression studies, indicating sharing of downstream cis-/trans-gene regulation across tissues and CMDs. In contrast, the regulatory effects of other GWAS risk SNPs were tissue-specific; abdominal fat emerged as an important gene-regulatory site for blood lipids, such as for the low-density lipoprotein cholesterol and coronary artery disease risk gene PCSK9 STARNET provides insights into gene-regulatory mechanisms for CMD risk loci, facilitating their translation into opportunities for diagnosis, therapy, and prevention.
Collapse
Affiliation(s)
- Oscar Franzén
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden
| | - Raili Ermel
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Department of Cardiac Surgery, Tartu University Hospital, 1a Ludwig Puusepa Street, 50406 Tartu, Estonia
| | - Ariella Cohain
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Nicholas K Akers
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Husain A Talukdar
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Hassan Foroughi-Asl
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Claudia Giambartolomei
- Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - John F Fullard
- Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Katyayani Sukhavasi
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia
| | - Sulev Köks
- Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia
| | - Li-Ming Gan
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Chiara Giannarelli
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Cardiovascular Research Center Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Jason C Kovacic
- Cardiovascular Research Center Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Christer Betsholtz
- AstraZeneca-Karolinska Integrated CardioMetabolic Centre (ICMC), Karolinska Institutet, Novum, Blickagången 6, 141 57 Huddinge, Sweden. Department of Immunology, Genetics and Pathology Dag Hammarskjölds Väg 20, 751 85 Uppsala, Sweden
| | - Bojan Losic
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Tom Michoel
- Division of Genetics and Genomics, The Roslin Institute, University of Edinburgh, Old College, South Bridge, Edinburgh EH8 9YL, UK
| | - Ke Hao
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Division of Psychiatric Genomics, Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Department of Psychiatry, J. J. Peters VA Medical Center, Mental Illness Research Education and Clinical Center (MIRECC), 130 West Kingsbridge Road, Bronx, NY 10468, USA
| | - Josefin Skogsberg
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden
| | - Arno Ruusalepp
- Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden. Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Department of Cardiac Surgery, Tartu University Hospital, 1a Ludwig Puusepa Street, 50406 Tartu, Estonia
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York , NY 10029, USA. Clinical Gene Networks AB, Jungfrugatan 10, 114 44 Stockholm, Sweden. Department of Pathophysiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Biomeedikum, Ravila 19, 50411, Tartu, Estonia. Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, 171 77 Stockholm, Sweden.
| |
Collapse
|
424
|
Novel Grb14-Mediated Cross Talk between Insulin and p62/Nrf2 Pathways Regulates Liver Lipogenesis and Selective Insulin Resistance. Mol Cell Biol 2016; 36:2168-81. [PMID: 27215388 DOI: 10.1128/mcb.00170-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/17/2016] [Indexed: 12/24/2022] Open
Abstract
A long-standing paradox in the pathophysiology of metabolic diseases is the selective insulin resistance of the liver. It is characterized by a blunted action of insulin to reduce glucose production, contributing to hyperglycemia, while de novo lipogenesis remains insulin sensitive, participating in turn to hepatic steatosis onset. The underlying molecular bases of this conundrum are not yet fully understood. Here, we established a model of selective insulin resistance in mice by silencing an inhibitor of insulin receptor catalytic activity, the growth factor receptor binding protein 14 (Grb14) in liver. Indeed, Grb14 knockdown enhanced hepatic insulin signaling but also dramatically inhibited de novo fatty acid synthesis. In the liver of obese and insulin-resistant mice, downregulation of Grb14 markedly decreased blood glucose and improved liver steatosis. Mechanistic analyses showed that upon Grb14 knockdown, the release of p62/sqstm1, a partner of Grb14, activated the transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2), which in turn repressed the lipogenic nuclear liver X receptor (LXR). Our study reveals that Grb14 acts as a new signaling node that regulates lipogenesis and modulates insulin sensitivity in the liver by acting at a crossroad between the insulin receptor and the p62-Nrf2-LXR signaling pathways.
Collapse
|
425
|
Adefurin A, Vanderbilt C, Okafor C, Kawai V, Li C, Shah A, Wei WQ, Kurnik D, Stein CM. Alpha2A adrenergic receptor genetic variation contributes to hyperglycemia after myocardial infarction. Int J Cardiol 2016; 215:482-6. [PMID: 27131769 PMCID: PMC4879094 DOI: 10.1016/j.ijcard.2016.04.079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/11/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is frequently associated with transient hyperglycemia even in patients without pre-existing diabetes. Acute stress can lead to increased blood glucose through the effect of catecholamines on alpha2A-adrenergic receptors (α2A-ARs) present in pancreatic islet β-cells. Variation in the gene (ADRA2A) that encodes the α2A-AR affects insulin release and glucose control and may play a particularly important role during times of stress. METHODS We performed a retrospective cohort study using de-identified electronic medical records linked to a DNA repository in 521 Caucasians and 55 African-American non-diabetic patients with AMI. We examined the association between admission blood glucose concentrations and ten selected ADRA2A SNPs in Caucasians. RESULTS Three ADRA2A SNPS were associated with stress-induced hyperglycemia in Caucasians. Individuals homozygous for the rs10885122 variant (n=9) had a 23% lower admission glucose (geometric mean [95% CI], 99 [83-118]mg/dl) compared with non-carriers (121 [118-125] mg/dl; n=401; P=0.001). Admission glucose was 14% higher in rs1800544 variant homozygotes (134 [119-150]mg/dl; n=36) compared to non-carriers (118 [115-121]mg/dl; n=290, P=0.046). Furthermore, homozygotes of the rs553668 variant (n=13) had a 13% higher glucose (133 [110-160]mg/dl) compared to non-carriers (118 [115-122]mg/dl; n=366; P=0.056). Haplotypes including these ADRA2A SNPs were associated with higher admission glucose levels. CONCLUSIONS Three ADRA2A genetic variants are associated with blood glucose and stress-induced hyperglycemia after AMI in Caucasians.
Collapse
Affiliation(s)
- Abiodun Adefurin
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Charles Vanderbilt
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Chimalum Okafor
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Vivian Kawai
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Chun Li
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Anushi Shah
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Daniel Kurnik
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA; Clinical Pharmacology Unit, Rambam Medical Center, Haifa, Israel; Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| | - C Michael Stein
- Departments of Medicine and Pharmacology, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
426
|
Abstract
As with other complex diseases, unbiased association studies followed by physiological and experimental characterization have for years formed a paradigm for identifying genes or processes of relevance to type 2 diabetes mellitus (T2D). Recent large-scale common and rare variant genome-wide association studies (GWAS) suggest that substantially larger association studies are needed to identify most T2D loci in the population. To hasten clinical translation of genetic discoveries, new paradigms are also required to aid specialized investigation of nascent hypotheses. We argue for an integrated T2D knowledgebase, designed for a worldwide community to access aggregated large-scale genetic data sets, as one paradigm to catalyse convergence of these efforts.
Collapse
|
427
|
ForestPMPlot: A Flexible Tool for Visualizing Heterogeneity Between Studies in Meta-analysis. G3-GENES GENOMES GENETICS 2016; 6:1793-8. [PMID: 27194809 PMCID: PMC4938634 DOI: 10.1534/g3.116.029439] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Meta-analysis has become a popular tool for genetic association studies to combine different genetic studies. A key challenge in meta-analysis is heterogeneity, or the differences in effect sizes between studies. Heterogeneity complicates the interpretation of meta-analyses. In this paper, we describe ForestPMPlot, a flexible visualization tool for analyzing studies included in a meta-analysis. The main feature of the tool is visualizing the differences in the effect sizes of the studies to understand why the studies exhibit heterogeneity for a particular phenotype and locus pair under different conditions. We show the application of this tool to interpret a meta-analysis of 17 mouse studies, and to interpret a multi-tissue eQTL study.
Collapse
|
428
|
Liu CT, Raghavan S, Maruthur N, Kabagambe EK, Hong J, Ng MCY, Hivert MF, Lu Y, An P, Bentley AR, Drolet AM, Gaulton KJ, Guo X, Armstrong LL, Irvin MR, Li M, Lipovich L, Rybin DV, Taylor KD, Agyemang C, Palmer ND, Cade BE, Chen WM, Dauriz M, Delaney JAC, Edwards TL, Evans DS, Evans MK, Lange LA, Leong A, Liu J, Liu Y, Nayak U, Patel SR, Porneala BC, Rasmussen-Torvik LJ, Snijder MB, Stallings SC, Tanaka T, Yanek LR, Zhao W, Becker DM, Bielak LF, Biggs ML, Bottinger EP, Bowden DW, Chen G, Correa A, Couper DJ, Crawford DC, Cushman M, Eicher JD, Fornage M, Franceschini N, Fu YP, Goodarzi MO, Gottesman O, Hara K, Harris TB, Jensen RA, Johnson AD, Jhun MA, Karter AJ, Keller MF, Kho AN, Kizer JR, Krauss RM, Langefeld CD, Li X, Liang J, Liu S, Lowe WL, Mosley TH, North KE, Pacheco JA, Peyser PA, Patrick AL, Rice KM, Selvin E, Sims M, Smith JA, Tajuddin SM, Vaidya D, Wren MP, Yao J, Zhu X, Ziegler JT, Zmuda JM, Zonderman AB, Zwinderman AH, Adeyemo A, Boerwinkle E, Ferrucci L, Hayes MG, Kardia SLR, Miljkovic I, Pankow JS, Rotimi CN, Sale MM, Wagenknecht LE, et alLiu CT, Raghavan S, Maruthur N, Kabagambe EK, Hong J, Ng MCY, Hivert MF, Lu Y, An P, Bentley AR, Drolet AM, Gaulton KJ, Guo X, Armstrong LL, Irvin MR, Li M, Lipovich L, Rybin DV, Taylor KD, Agyemang C, Palmer ND, Cade BE, Chen WM, Dauriz M, Delaney JAC, Edwards TL, Evans DS, Evans MK, Lange LA, Leong A, Liu J, Liu Y, Nayak U, Patel SR, Porneala BC, Rasmussen-Torvik LJ, Snijder MB, Stallings SC, Tanaka T, Yanek LR, Zhao W, Becker DM, Bielak LF, Biggs ML, Bottinger EP, Bowden DW, Chen G, Correa A, Couper DJ, Crawford DC, Cushman M, Eicher JD, Fornage M, Franceschini N, Fu YP, Goodarzi MO, Gottesman O, Hara K, Harris TB, Jensen RA, Johnson AD, Jhun MA, Karter AJ, Keller MF, Kho AN, Kizer JR, Krauss RM, Langefeld CD, Li X, Liang J, Liu S, Lowe WL, Mosley TH, North KE, Pacheco JA, Peyser PA, Patrick AL, Rice KM, Selvin E, Sims M, Smith JA, Tajuddin SM, Vaidya D, Wren MP, Yao J, Zhu X, Ziegler JT, Zmuda JM, Zonderman AB, Zwinderman AH, Adeyemo A, Boerwinkle E, Ferrucci L, Hayes MG, Kardia SLR, Miljkovic I, Pankow JS, Rotimi CN, Sale MM, Wagenknecht LE, Arnett DK, Chen YDI, Nalls MA, Province MA, Kao WHL, Siscovick DS, Psaty BM, Wilson JG, Loos RJF, Dupuis J, Rich SS, Florez JC, Rotter JI, Morris AP, Meigs JB. Trans-ethnic Meta-analysis and Functional Annotation Illuminates the Genetic Architecture of Fasting Glucose and Insulin. Am J Hum Genet 2016; 99:56-75. [PMID: 27321945 PMCID: PMC5005440 DOI: 10.1016/j.ajhg.2016.05.006] [Show More Authors] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022] Open
Abstract
Knowledge of the genetic basis of the type 2 diabetes (T2D)-related quantitative traits fasting glucose (FG) and insulin (FI) in African ancestry (AA) individuals has been limited. In non-diabetic subjects of AA (n = 20,209) and European ancestry (EA; n = 57,292), we performed trans-ethnic (AA+EA) fine-mapping of 54 established EA FG or FI loci with detailed functional annotation, assessed their relevance in AA individuals, and sought previously undescribed loci through trans-ethnic (AA+EA) meta-analysis. We narrowed credible sets of variants driving association signals for 22/54 EA-associated loci; 18/22 credible sets overlapped with active islet-specific enhancers or transcription factor (TF) binding sites, and 21/22 contained at least one TF motif. Of the 54 EA-associated loci, 23 were shared between EA and AA. Replication with an additional 10,096 AA individuals identified two previously undescribed FI loci, chrX FAM133A (rs213676) and chr5 PELO (rs6450057). Trans-ethnic analyses with regulatory annotation illuminate the genetic architecture of glycemic traits and suggest gene regulation as a target to advance precision medicine for T2D. Our approach to utilize state-of-the-art functional annotation and implement trans-ethnic association analysis for discovery and fine-mapping offers a framework for further follow-up and characterization of GWAS signals of complex trait loci.
Collapse
Affiliation(s)
- Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118, USA.
| | - Sridharan Raghavan
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Veterans Affairs Medical Center, Eastern Colorado Health Care System, Denver, CO 80220, USA; Division of General Internal Medicine, Department of Medicine, University of Colorado School of Medicine, Denver, CO 80220, USA
| | - Nisa Maruthur
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD 21287, USA; Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Edmond Kato Kabagambe
- Division of Epidemiology, Department of Medicine, School of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Jaeyoung Hong
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118, USA
| | - Maggie C Y Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Center for Diabetes Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Universite de Sherbrooke, Sherbrooke, QC J1G 0A2, Canada
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, School of Medicine, Washington University, St Louis, MO 63108, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Anne M Drolet
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Kyle J Gaulton
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Loren L Armstrong
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marguerite R Irvin
- Department of Epidemiology, School of Public Health, University of Alabama - Birmingham, Birmingham, AL 35294, USA
| | - Man Li
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Leonard Lipovich
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Neurology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Denis V Rybin
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Charles Agyemang
- Department of Public Health, Academic Medical Center Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Nicholette D Palmer
- Center for Genomics and Personalized Medicine Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei-Min Chen
- Center for Public Health Genomics, Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Marco Dauriz
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Joseph A C Delaney
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, School of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Leslie A Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27607, USA
| | - Aaron Leong
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jingmin Liu
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Uma Nayak
- Center for Public Health Genomics, Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Sanjay R Patel
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bianca C Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marieke B Snijder
- Department of Public Health, Academic Medical Center Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Sarah C Stallings
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute of Aging at Harbor Hospital, Baltimore, MD 21225, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diane M Becker
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; Department of Health Policy and Management, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary L Biggs
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA; Cardiovascular Health Research Unit, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Center for Diabetes Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - David J Couper
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Dana C Crawford
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mary Cushman
- Department of Medicine and Pathology, University of Vermont, College of Medicine, Burlington, VT 05405, USA
| | - John D Eicher
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Framingham, MA 01702, USA
| | - Myriam Fornage
- Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Yi-Ping Fu
- Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung, and Blood Institute, NIH, Framingham, MA 01702, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes & Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kazuo Hara
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Diabetes, Endocrinology, and Metabolism, Tokyo Medical University, Tokyo 163-0023, Japan
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, NIH, Bethesda, MD 20892, USA
| | - Richard A Jensen
- Cardiovascular Health Research Unit, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Andrew D Johnson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Framingham, MA 01702, USA
| | - Min A Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew J Karter
- Division of Research, Kaiser Permanente, Northern California Region, Oakland, CA 94612, USA
| | - Margaux F Keller
- Department of Genetics and Pharmacogenomics, Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Abel N Kho
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ronald M Krauss
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Carl D Langefeld
- Center for Public Health Genomics, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jingling Liang
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Simin Liu
- Department of Epidemiology, Brown University, Providence, RI 02912, USA; Department of Medicine, Brown University, Providence, RI 02903, USA
| | - William L Lowe
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thomas H Mosley
- Division of Geriatrics/Gerontology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Jennifer A Pacheco
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alan L Patrick
- Tobago Health Studies Office, Scarborough, Tobago, Trinidad and Tobago
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth Selvin
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD 21287, USA; Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Mario Sims
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Dhananjay Vaidya
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA; GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Mary P Wren
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Julie T Ziegler
- Center for Public Health Genomics, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Joseph M Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology & Population Science, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD 21224, US
| | - Aeilko H Zwinderman
- Department of Public Health, Academic Medical Center Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Eric Boerwinkle
- Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute of Aging at Harbor Hospital, Baltimore, MD 21225, USA
| | - M Geoffrey Hayes
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Michele M Sale
- Center for Public Health Genomics, Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Donna K Arnett
- University of Kentucky College of Public Health, Lexington, KY 40563, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, School of Medicine, Washington University, St Louis, MO 63108, USA
| | - W H Linda Kao
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - David S Siscovick
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Cardiovascular Health Research Unit, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA; The New York Academy of Medicine, New York, NY 10029, USA
| | - Bruce M Psaty
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Cardiovascular Health Research Unit, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Health Services, University of Washington, Seattle, WA 98195, USA; Group Health Research Institute, Group Health Cooperative, Seattle, WA 98101, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josée Dupuis
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02118, USA; National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
| | - Stephen S Rich
- Center for Public Health Genomics, Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jose C Florez
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Institute of Translational Medicine, Department of Biostatistics, University of Liverpool, Liverpool L69 3BX, UK
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
429
|
Aschard H. A perspective on interaction effects in genetic association studies. Genet Epidemiol 2016; 40:678-688. [PMID: 27390122 PMCID: PMC5132101 DOI: 10.1002/gepi.21989] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/20/2016] [Accepted: 06/05/2016] [Indexed: 11/29/2022]
Abstract
The identification of gene–gene and gene–environment interaction in human traits and diseases is an active area of research that generates high expectation, and most often lead to high disappointment. This is partly explained by a misunderstanding of the inherent characteristics of standard regression‐based interaction analyses. Here, I revisit and untangle major theoretical aspects of interaction tests in the special case of linear regression; in particular, I discuss variables coding scheme, interpretation of effect estimate, statistical power, and estimation of variance explained in regard of various hypothetical interaction patterns. Linking this components it appears first that the simplest biological interaction models—in which the magnitude of a genetic effect depends on a common exposure—are among the most difficult to identify. Second, I highlight the demerit of the current strategy to evaluate the contribution of interaction effects to the variance of quantitative outcomes and argue for the use of new approaches to overcome this issue. Finally, I explore the advantages and limitations of multivariate interaction models, when testing for interaction between multiple SNPs and/or multiple exposures, over univariate approaches. Together, these new insights can be leveraged for future method development and to improve our understanding of the genetic architecture of multifactorial traits.
Collapse
Affiliation(s)
- Hugues Aschard
- Department of Epidemiology, Harvard T.H. School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
430
|
Jung SY, Sobel EM, Papp JC, Crandall CJ, Fu AN, Zhang ZF. Obesity and associated lifestyles modify the effect of glucose metabolism-related genetic variants on impaired glucose homeostasis among postmenopausal women. Genet Epidemiol 2016; 40:520-30. [PMID: 27377425 DOI: 10.1002/gepi.21991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/24/2016] [Accepted: 06/05/2016] [Indexed: 01/19/2023]
Abstract
PURPOSE Impaired glucose metabolism-related genetic variants likely interact with obesity-modifiable factors in response to glucose intolerance, yet their interconnected pathways have not been fully characterized. METHODS With data from 1,027 postmenopausal participants of the Genomics and Randomized Trials Network study and 15 single-nucleotide polymorphisms (SNPs) associated with glucose homeostasis, we assessed whether obesity, physical activity, and high dietary fat intake interact with the SNP-glucose variations. We used regression analysis plus stratification and graphic approaches. RESULTS Across carriers of the 15 SNPs, fasting levels of glucose, insulin, and homeostatic model assessment-insulin resistance (HOMA-IR) were higher in obese, inactive, and high fat-diet women than in their respective counterparts. Carriers within subgroups differently demonstrated the direction and/or magnitude of the variants' effect on glucose-relevant traits. Variants in GCKR, GCK, DGKB/TMEM195 (P for interactions = 0.02, 0.02, and 0.01), especially, showed interactions with obesity: obese, inactive, and high fat-diet women had greater increases in fasting glucose, insulin, and HOMA-IR levels. Obese carriers at TCF7L2 variant had greater increases in fasting glucose levels than nonobese carriers (P for interaction = 0.04), whereas active women had greater decreases in insulin and HOMA-IR levels than inactive women (P for interaction = 0.02 in both levels). CONCLUSIONS Our data support the important role of obesity in modifying glucose homeostasis in response to glucose metabolism-relevant variants. These findings may inform research on the role of glucose homeostasis in the etiology of chronic disease and the development of intervention strategies to reduce risk in postmenopausal women.
Collapse
Affiliation(s)
- Su Yon Jung
- Translational Sciences Section, Jonsson Comprehensive Cancer Center, School of Nursing, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Eric M Sobel
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jeanette C Papp
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Carolyn J Crandall
- Division of General Internal Medicine, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Alan N Fu
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Zuo-Feng Zhang
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
431
|
Tin A, Balakrishnan P, Beaty TH, Boerwinkle E, Hoogeveen RC, Young JH, Kao WHL. GCKR and PPP1R3B identified as genome-wide significant loci for plasma lactate: the Atherosclerosis Risk in Communities (ARIC) study. Diabet Med 2016; 33:968-75. [PMID: 26433129 PMCID: PMC4819009 DOI: 10.1111/dme.12971] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2015] [Indexed: 12/22/2022]
Abstract
AIM To investigate the genetic influence of circulating lactate level, a marker of oxidative capacity associated with diabetes. METHODS We conducted a genome-wide association study of log-transformed plasma lactate levels in 6901 European-American participants in the Atherosclerosis Risk in Communities study. For regions that achieved genome-wide significance in European-American participants, we conducted candidate region analysis in African-American subjects and tested for interaction between metformin use and the index single nucleotide polymorphisms for plasma lactate in European-American subjects. RESULTS The genome-wide association study in European-American subjects identified two genome-wide significant loci, GCKR (rs1260326, T allele β=0.08; P=1.8×10(-47) ) and PPP1R3B/LOC157273 (rs9987289, A allele β=0.06; P=1.6×10(-9) ). The index single nucleotide polymorphisms in these two loci explain 3.3% of the variance in log-transformed plasma lactate levels among the European-American subjects. In the African-American subjects, based on a region-significant threshold, the index single nucleotide polymorphism at GCKR was associated with plasma lactate but that at PPP1R3B/LOC157273 was not. Metformin use appeared to strengthen the association between the index single nucleotide polymorphism at PPP1R3B/LOC157273 and plasma lactate in European-American subjects (P for interaction=0.01). CONCLUSIONS We identified GCKR and PPP1R3B/LOC157273 as two genome-wide significant loci of plasma lactate. Both loci are associated with other diabetes-related phenotypes. These findings increase our understanding of the genetic control of lactate metabolism.
Collapse
Affiliation(s)
- A Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - P Balakrishnan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - T H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - E Boerwinkle
- Human Genetics Center, University of Texas School of Public Health, Houston, TX, USA
| | - R C Hoogeveen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - J H Young
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - W H L Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
432
|
Pickrell JK, Berisa T, Liu JZ, Ségurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet 2016; 48:709-17. [PMID: 27182965 PMCID: PMC5207801 DOI: 10.1038/ng.3570] [Citation(s) in RCA: 792] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 04/20/2016] [Indexed: 12/14/2022]
Abstract
We performed a scan for genetic variants associated with multiple phenotypes by comparing large genome-wide association studies (GWAS) of 42 traits or diseases. We identified 341 loci (at a false discovery rate of 10%) associated with multiple traits. Several loci are associated with multiple phenotypes; for example, a nonsynonymous variant in the zinc transporter SLC39A8 influences seven of the traits, including risk of schizophrenia (rs13107325: log-transformed odds ratio (log OR) = 0.15, P = 2 × 10(-12)) and Parkinson disease (log OR = -0.15, P = 1.6 × 10(-7)), among others. Second, we used these loci to identify traits that have multiple genetic causes in common. For example, variants associated with increased risk of schizophrenia also tended to be associated with increased risk of inflammatory bowel disease. Finally, we developed a method to identify pairs of traits that show evidence of a causal relationship. For example, we show evidence that increased body mass index causally increases triglyceride levels.
Collapse
Affiliation(s)
- Joseph K Pickrell
- New York Genome Center, New York, New York, USA
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | | | - Jimmy Z Liu
- New York Genome Center, New York, New York, USA
| | - Laure Ségurel
- UMR 7206 Eco-Anthropologie et Ethnobiologie, CNRS, MNHN, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | |
Collapse
|
433
|
Gan W, Walters RG, Holmes MV, Bragg F, Millwood IY, Banasik K, Chen Y, Du H, Iona A, Mahajan A, Yang L, Bian Z, Guo Y, Clarke RJ, Li L, McCarthy MI, Chen Z. Evaluation of type 2 diabetes genetic risk variants in Chinese adults: findings from 93,000 individuals from the China Kadoorie Biobank. Diabetologia 2016; 59:1446-1457. [PMID: 27053236 PMCID: PMC4901105 DOI: 10.1007/s00125-016-3920-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/22/2016] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS Genome-wide association studies (GWAS) have discovered many risk variants for type 2 diabetes. However, estimates of the contributions of risk variants to type 2 diabetes predisposition are often based on highly selected case-control samples, and reliable estimates of population-level effect sizes are missing, especially in non-European populations. METHODS The individual and cumulative effects of 59 established type 2 diabetes risk loci were measured in a population-based China Kadoorie Biobank (CKB) study of 93,000 Chinese adults, including >7,100 diabetes cases. RESULTS Association signals were directionally consistent between CKB and the original discovery GWAS: of 56 variants passing quality control, 48 showed the same direction of effect (binomial test, p = 2.3 × 10(-8)). We observed a consistent overall trend towards lower risk variant effect sizes in CKB than in case-control samples of GWAS meta-analyses (mean 19-22% decrease in log odds, p ≤ 0.0048), likely to reflect correction of both 'winner's curse' and spectrum bias effects. The association with risk of diabetes of a genetic risk score, based on lead variants at 25 loci considered to act through beta cell function, demonstrated significant interactions with several measures of adiposity (BMI, waist circumference [WC], WHR and percentage body fat [PBF]; all p interaction < 1 × 10(-4)), with a greater effect being observed in leaner adults. CONCLUSIONS/INTERPRETATION Our study provides further evidence of shared genetic architecture for type 2 diabetes between Europeans and East Asians. It also indicates that even very large GWAS meta-analyses may be vulnerable to substantial inflation of effect size estimates, compared with those observed in large-scale population-based cohort studies. ACCESS TO RESEARCH MATERIALS Details of how to access China Kadoorie Biobank data and details of the data release schedule are available from www.ckbiobank.org/site/Data+Access .
Collapse
Affiliation(s)
- Wei Gan
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital Campus, Old Road, Headington, Oxford, OX3 7LJ, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Michael V Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Fiona Bragg
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Karina Banasik
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital Campus, Old Road, Headington, Oxford, OX3 7LJ, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen, Denmark
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Andri Iona
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital Campus, Old Road, Headington, Oxford, OX3 7LJ, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, People's Republic of China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, People's Republic of China
| | - Robert J Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Liming Li
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, People's Republic of China
- School of Public Health, Peking University Health Sciences Center, Beijing, People's Republic of China
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital Campus, Old Road, Headington, Oxford, OX3 7LJ, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
- National Institute of Health Research Oxford Biomedical Research Centre, Oxford, UK.
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK.
| | | |
Collapse
|
434
|
Abstract
The genome is often the conduit through which environmental exposures convey their effects on health and disease. Whilst not all diseases act by directly perturbing the genome, the phenotypic responses are often genetically determined. Hence, whilst diseases are often defined has having differing degrees of genetic determination, genetic and environmental factors are, with few exceptions, inseparable features of most diseases, not least type 2 diabetes. It follows that to optimize diabetes, prevention and treatment will require that the etiological roles of genetic and environmental risk factors be jointly considered. As we discuss here, studies focused on quantifying gene-environment and gene-treatment interactions are gathering momentum and may eventually yield data that helps guide health-related choices and medical interventions for type 2 diabetes and other complex diseases.
Collapse
Affiliation(s)
- Paul W Franks
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Center, Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital Malmö, Lund University, Building 91, Level 10, Jan Waldenströms gata 35, 205 02, Malmö, Sweden.
- Department of Public Health and Clinical Medicine, Umeå University, 90188, Umeå, Sweden.
- Department of Nutrition, Harvard School of Public Health, Boston, MA, 02115, USA.
| | - Guillaume Paré
- Population Health Research Institute, McMaster University, Hamilton General Hospital Campus, DB-CVSRI, 237 Barton Street East, Room C3103, Hamilton, ON, L8L 2X2, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Department of Clinical Epidemiology and Biostatistics, Population Genomics Program, McMaster University, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| |
Collapse
|
435
|
Muka T, Nano J, Voortman T, Braun KVE, Ligthart S, Stranges S, Bramer WM, Troup J, Chowdhury R, Dehghan A, Franco OH. The role of global and regional DNA methylation and histone modifications in glycemic traits and type 2 diabetes: A systematic review. Nutr Metab Cardiovasc Dis 2016; 26:553-566. [PMID: 27146363 DOI: 10.1016/j.numecd.2016.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 04/04/2016] [Accepted: 04/04/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND New evidence suggests the potential involvement of epigenetic mechanisms in type 2 diabetes (T2D) as a crucial interface between the effects of genetic predisposition and environmental influences. AIM To systematically review studies investigating the association between epigenetic marks (DNA methylation and histone modifications) with T2D and glycemic traits (glucose and insulin levels, insulin resistance measured by HOMA-IR). METHOD AND RESULTS Six bibliographic databases (Embase.com, Medline (Ovid), Web-of-Science, PubMed, Cochrane Central and Google Scholar) were screened until 28th August 2015. We included randomized controlled trials, cohort, case-control and cross-sectional studies in humans that examined the association between epigenetic marks (global, candidate or genome-wide methylation of DNA and histone modifications) with T2D, glucose and insulin levels and insulin metabolism. Of the initially identified 3879 references, 53 articles, based on 47 unique studies met our inclusion criteria. Overall, data were available on 10,823 participants, with a total of 3358 T2D cases. There was no consistent evidence for an association between global DNA-methylation with T2D, glucose, insulin and insulin resistance. The studies reported epigenetic regulation of several candidate genes for diabetes susceptibility in blood cells, muscle, adipose tissue and placenta to be related with T2D without any general overlap between them. Histone modifications in relation to T2D were reported only in 3 observational studies. CONCLUSIONS AND RELEVANCE Current evidence supports an association between epigenetic marks and T2D. However, overall evidence is limited, highlighting the need for further larger-scale and prospective investigations to establish whether epigenetic marks may influence the risk of developing T2D.
Collapse
Affiliation(s)
- T Muka
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - J Nano
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - T Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - K V E Braun
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - S Ligthart
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - S Stranges
- Department of Population Health, Luxembourg Institute of Health, Luxembourg
| | - W M Bramer
- Medical Library, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - J Troup
- Research and Development, Metagenics, Inc, USA
| | - R Chowdhury
- Department of Public Health & Primary Care, Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, United Kingdom
| | - A Dehghan
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - O H Franco
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
436
|
A Genome-Wide mQTL Analysis in Human Adipose Tissue Identifies Genetic Variants Associated with DNA Methylation, Gene Expression and Metabolic Traits. PLoS One 2016; 11:e0157776. [PMID: 27322064 PMCID: PMC4913906 DOI: 10.1371/journal.pone.0157776] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/03/2016] [Indexed: 01/17/2023] Open
Abstract
Little is known about the extent to which interactions between genetics and epigenetics may affect the risk of complex metabolic diseases and/or their intermediary phenotypes. We performed a genome-wide DNA methylation quantitative trait locus (mQTL) analysis in human adipose tissue of 119 men, where 592,794 single nucleotide polymorphisms (SNPs) were related to DNA methylation of 477,891 CpG sites, covering 99% of RefSeq genes. SNPs in significant mQTLs were further related to gene expression in adipose tissue and obesity related traits. We found 101,911 SNP-CpG pairs (mQTLs) in cis and 5,342 SNP-CpG pairs in trans showing significant associations between genotype and DNA methylation in adipose tissue after correction for multiple testing, where cis is defined as distance less than 500 kb between a SNP and CpG site. These mQTLs include reported obesity, lipid and type 2 diabetes loci, e.g. ADCY3/POMC, APOA5, CETP, FADS2, GCKR, SORT1 and LEPR. Significant mQTLs were overrepresented in intergenic regions meanwhile underrepresented in promoter regions and CpG islands. We further identified 635 SNPs in significant cis-mQTLs associated with expression of 86 genes in adipose tissue including CHRNA5, G6PC2, GPX7, RPL27A, THNSL2 and ZFP57. SNPs in significant mQTLs were also associated with body mass index (BMI), lipid traits and glucose and insulin levels in our study cohort and public available consortia data. Importantly, the Causal Inference Test (CIT) demonstrates how genetic variants mediate their effects on metabolic traits (e.g. BMI, cholesterol, high-density lipoprotein (HDL), hemoglobin A1c (HbA1c) and homeostatic model assessment of insulin resistance (HOMA-IR)) via altered DNA methylation in human adipose tissue. This study identifies genome-wide interactions between genetic and epigenetic variation in both cis and trans positions influencing gene expression in adipose tissue and in vivo (dys)metabolic traits associated with the development of obesity and diabetes.
Collapse
|
437
|
Karanth S, Zinkhan EK, Hill JT, Yost HJ, Schlegel A. FOXN3 Regulates Hepatic Glucose Utilization. Cell Rep 2016; 15:2745-55. [PMID: 27292639 DOI: 10.1016/j.celrep.2016.05.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/27/2016] [Accepted: 05/13/2016] [Indexed: 12/17/2022] Open
Abstract
A SNP (rs8004664) in the first intron of the FOXN3 gene is associated with human fasting blood glucose. We find that carriers of the risk allele have higher hepatic expression of the transcriptional repressor FOXN3. Rat Foxn3 protein and zebrafish foxn3 transcripts are downregulated during fasting, a process recapitulated in human HepG2 hepatoma cells. Transgenic overexpression of zebrafish foxn3 or human FOXN3 increases zebrafish hepatic gluconeogenic gene expression, whole-larval free glucose, and adult fasting blood glucose and also decreases expression of glycolytic genes. Hepatic FOXN3 overexpression suppresses expression of mycb, whose ortholog MYC is known to directly stimulate expression of glucose-utilization enzymes. Carriers of the rs8004664 risk allele have decreased MYC transcript abundance. Human FOXN3 binds DNA sequences in the human MYC and zebrafish mycb loci. We conclude that the rs8004664 risk allele drives excessive expression of FOXN3 during fasting and that FOXN3 regulates fasting blood glucose.
Collapse
Affiliation(s)
- Santhosh Karanth
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Erin K Zinkhan
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Jonathon T Hill
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - H Joseph Yost
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Amnon Schlegel
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
438
|
Wiemerslage L, Gohel PA, Maestri G, Hilmarsson TG, Mickael M, Fredriksson R, Williams MJ, Schiöth HB. The Drosophila ortholog of TMEM18 regulates insulin and glucagon-like signaling. J Endocrinol 2016; 229:233-43. [PMID: 27029472 DOI: 10.1530/joe-16-0040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 12/19/2022]
Abstract
Transmembrane protein 18 (TMEM18) is an ill-described, obesity-related gene, but few studies have explored its molecular function. We found single-nucleotide polymorphism data, suggesting that TMEM18 may be involved in the regulation/physiology of metabolic syndrome based on associations with insulin, homeostatic model assessment-β (HOMAβ), triglycerides, and blood sugar. We then found an ortholog in the Drosophila genome, knocked down Drosophila Tmem18 specifically in insulin-producing cells, and tested for its effects on metabolic function. Our results suggest that TMEM18 affects substrate levels through insulin and glucagon signaling, and its downregulation induces a metabolic state resembling type 2 diabetes. This work is the first to experimentally describe the metabolic consequences of TMEM18 knockdown, and further supports its association with obesity.
Collapse
Affiliation(s)
- Lyle Wiemerslage
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Priya A Gohel
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Giulia Maestri
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Torfi G Hilmarsson
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Michel Mickael
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Michael J Williams
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of NeuroscienceFunctional Pharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
439
|
Kong X, Xing X, Hong J, Zhang X, Yang W. Genetic variants associated with lean and obese type 2 diabetes in a Han Chinese population: A case-control study. Medicine (Baltimore) 2016; 95:e3841. [PMID: 27281091 PMCID: PMC4907669 DOI: 10.1097/md.0000000000003841] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Type 2 diabetes (T2D) is highly phenotypically heterogeneous. Genetics of the heterogeneity of lean and obese T2D is not clear. The aim of the present study was to identify the associations of T2D-related genetic variants with the risks for lean and obese T2D among the Chinese Han population. A case-control study consisting of 5338 T2D patients and 4663 normal glycemic controls of Chinese Han recruited in the Chinese National Diabetes and Metabolic Disorders Study was conducted. T2D cases were identified according to the 1999 World Health Organization criteria. Lean T2D was defined as T2D patient with a body mass index (BMI) <23 kg/m, whereas obese T2D was defined as T2D patient with a BMI ≥28 kg/m. Twenty-five genome-wide association studies previously validated T2D-related single-nucleotide polymorphisms (SNPs) were genotyped. A genotype risk score (GRS) based on the 25 SNPs was created. After adjusting for multiple covariates, SNPs in or near CDKAL1, CDKN2BAS, KCNQ1, TCF7L2, CDC123/CAMK1D, HHEX, and TCF2 were associated with the risk for lean T2D, and SNPs in or near KCNQ1 and FTO were associated with the risk for obese T2D. The results showed that the GRS for 25 T2D-related SNPs was more strongly associated with the risk for lean T2D (Ptrend = 2.66 × 10) than for obese T2D (Ptrend = 2.91 × 10) in our study population. Notably, the T2D GRS contributed to lower obesity-related measurements and greater β-cell dysfunction, including lower insulin levels in oral glucose tolerance test, decreased insulinogenic index, and Homeostasis Model Assessment for β-cell Function. In conclusion, our findings identified T2D-related genetic loci that contribute to the risk of lean and obese T2D individually and additively in a Chinese Han population. Moreover, the study highlights the contribution of known T2D genomic loci to the heterogeneity of lean and obese T2D in Chinese Hans.
Collapse
Affiliation(s)
| | | | | | | | - Wenying Yang
- ∗Correspondence: Wenying Yang, Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, P.R. China (e-mail: )
| |
Collapse
|
440
|
Scott RA, Freitag DF, Li L, Chu AY, Surendran P, Young R, Grarup N, Stancáková A, Chen Y, Varga TV, Yaghootkar H, Luan J, Zhao JH, Willems SM, Wessel J, Wang S, Maruthur N, Michailidou K, Pirie A, van der Lee SJ, Gillson C, Al Olama AA, Amouyel P, Arriola L, Arveiler D, Aviles-Olmos I, Balkau B, Barricarte A, Barroso I, Garcia SB, Bis JC, Blankenberg S, Boehnke M, Boeing H, Boerwinkle E, Borecki IB, Bork-Jensen J, Bowden S, Caldas C, Caslake M, Cupples LA, Cruchaga C, Czajkowski J, den Hoed M, Dunn JA, Earl HM, Ehret GB, Ferrannini E, Ferrieres J, Foltynie T, Ford I, Forouhi NG, Gianfagna F, Gonzalez C, Grioni S, Hiller L, Jansson JH, Jørgensen ME, Jukema JW, Kaaks R, Kee F, Kerrison ND, Key TJ, Kontto J, Kote-Jarai Z, Kraja AT, Kuulasmaa K, Kuusisto J, Linneberg A, Liu C, Marenne G, Mohlke KL, Morris AP, Muir K, Müller-Nurasyid M, Munroe PB, Navarro C, Nielsen SF, Nilsson PM, Nordestgaard BG, Packard CJ, Palli D, Panico S, Peloso GM, Perola M, Peters A, Poole CJ, Quirós JR, Rolandsson O, Sacerdote C, Salomaa V, Sánchez MJ, Sattar N, Sharp SJ, Sims R, Slimani N, Smith JA, Thompson DJ, Trompet S, Tumino R, et alScott RA, Freitag DF, Li L, Chu AY, Surendran P, Young R, Grarup N, Stancáková A, Chen Y, Varga TV, Yaghootkar H, Luan J, Zhao JH, Willems SM, Wessel J, Wang S, Maruthur N, Michailidou K, Pirie A, van der Lee SJ, Gillson C, Al Olama AA, Amouyel P, Arriola L, Arveiler D, Aviles-Olmos I, Balkau B, Barricarte A, Barroso I, Garcia SB, Bis JC, Blankenberg S, Boehnke M, Boeing H, Boerwinkle E, Borecki IB, Bork-Jensen J, Bowden S, Caldas C, Caslake M, Cupples LA, Cruchaga C, Czajkowski J, den Hoed M, Dunn JA, Earl HM, Ehret GB, Ferrannini E, Ferrieres J, Foltynie T, Ford I, Forouhi NG, Gianfagna F, Gonzalez C, Grioni S, Hiller L, Jansson JH, Jørgensen ME, Jukema JW, Kaaks R, Kee F, Kerrison ND, Key TJ, Kontto J, Kote-Jarai Z, Kraja AT, Kuulasmaa K, Kuusisto J, Linneberg A, Liu C, Marenne G, Mohlke KL, Morris AP, Muir K, Müller-Nurasyid M, Munroe PB, Navarro C, Nielsen SF, Nilsson PM, Nordestgaard BG, Packard CJ, Palli D, Panico S, Peloso GM, Perola M, Peters A, Poole CJ, Quirós JR, Rolandsson O, Sacerdote C, Salomaa V, Sánchez MJ, Sattar N, Sharp SJ, Sims R, Slimani N, Smith JA, Thompson DJ, Trompet S, Tumino R, van der A DL, van der Schouw YT, Virtamo J, Walker M, Walter K, Abraham JE, Amundadottir LT, Aponte JL, Butterworth AS, Dupuis J, Easton DF, Eeles RA, Erdmann J, Franks PW, Frayling TM, Hansen T, Howson JMM, Jørgensen T, Kooner J, Laakso M, Langenberg C, McCarthy MI, Pankow JS, Pedersen O, Riboli E, Rotter JI, Saleheen D, Samani NJ, Schunkert H, Vollenweider P, O'Rahilly S, Deloukas P, Danesh J, Goodarzi MO, Kathiresan S, Meigs JB, Ehm MG, Wareham NJ, Waterworth DM. A genomic approach to therapeutic target validation identifies a glucose-lowering GLP1R variant protective for coronary heart disease. Sci Transl Med 2016; 8:341ra76. [PMID: 27252175 PMCID: PMC5219001 DOI: 10.1126/scitranslmed.aad3744] [Show More Authors] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
Abstract
Regulatory authorities have indicated that new drugs to treat type 2 diabetes (T2D) should not be associated with an unacceptable increase in cardiovascular risk. Human genetics may be able to guide development of antidiabetic therapies by predicting cardiovascular and other health endpoints. We therefore investigated the association of variants in six genes that encode drug targets for obesity or T2D with a range of metabolic traits in up to 11,806 individuals by targeted exome sequencing and follow-up in 39,979 individuals by targeted genotyping, with additional in silico follow-up in consortia. We used these data to first compare associations of variants in genes encoding drug targets with the effects of pharmacological manipulation of those targets in clinical trials. We then tested the association of those variants with disease outcomes, including coronary heart disease, to predict cardiovascular safety of these agents. A low-frequency missense variant (Ala316Thr; rs10305492) in the gene encoding glucagon-like peptide-1 receptor (GLP1R), the target of GLP1R agonists, was associated with lower fasting glucose and T2D risk, consistent with GLP1R agonist therapies. The minor allele was also associated with protection against heart disease, thus providing evidence that GLP1R agonists are not likely to be associated with an unacceptable increase in cardiovascular risk. Our results provide an encouraging signal that these agents may be associated with benefit, a question currently being addressed in randomized controlled trials. Genetic variants associated with metabolic traits and multiple disease outcomes can be used to validate therapeutic targets at an early stage in the drug development process.
Collapse
Affiliation(s)
- Robert A Scott
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
| | - Daniel F Freitag
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK. The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Li Li
- Statistical Genetics, Projects, Clinical Platforms, and Sciences (PCPS), GlaxoSmithKline, Research Triangle Park, NC 27709, USA
| | - Audrey Y Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Praveen Surendran
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK
| | - Robin Young
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alena Stancáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Yuning Chen
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 Malmö, Sweden
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Jian'an Luan
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Jing Hua Zhao
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Sara M Willems
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK. Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, 3000 CE Rotterdam, Netherlands
| | - Jennifer Wessel
- Department of Epidemiology, Fairbanks School of Public Health, Indianapolis, IN 46202, USA. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shuai Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Nisa Maruthur
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD 21205, USA. Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kyriaki Michailidou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Ailith Pirie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Sven J van der Lee
- Department of Epidemiology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Christopher Gillson
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Philippe Amouyel
- University of Lille, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, UMR 1167, RID-AGE, F-59000 Lille, France
| | - Larraitz Arriola
- Public Health Division of Gipuzkoa, San Sebastian 20013, Spain. Instituto BIO-Donostia, Basque Government, San Sebastian 20014, Spain. CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain
| | - Dominique Arveiler
- Department of Epidemiology and Public Health (EA3430), University of Strasbourg, 67085 Strasbourg, France
| | - Iciar Aviles-Olmos
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Beverley Balkau
- INSERM, Centre de Recherche en Epidémiologie et Santé des Populations (CESP), 94807 Villejuif, France. Univeristy of Paris-Sud, F-94805 Villejuif, France
| | - Aurelio Barricarte
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain. Navarre Public Health Institute (ISPN), Pamplona 31003, Spain
| | - Inês Barroso
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK. University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Sara Benlloch Garcia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109-2029, USA
| | - Heiner Boeing
- German Institute of Human Nutrition, Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77025, USA. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ingrid B Borecki
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jette Bork-Jensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sarah Bowden
- Cancer Research UK Clinical Trials Unit, Institute for Cancer Studies, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA. Framingham Heart Study, National Heart, Lung, and Blood Institute (NHLBI), Framingham, MA 01702-5827, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jacek Czajkowski
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Marcel den Hoed
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, SE-752 37 Uppsala, Sweden
| | - Janet A Dunn
- Warwick Clinical Trials Unit, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Helena M Earl
- University of Cambridge and National Institute of Health Research Cambridge Biomedical Research Centre, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge CB2 0QQ, UK
| | - Georg B Ehret
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ele Ferrannini
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, 56124 Pisa, Italy
| | - Jean Ferrieres
- Department of Epidemiology, UMR 1027, INSERM, Centre Hospitalier Universitaire (CHU) de Toulouse, 31000 Toulouse, France
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Ian Ford
- University of Glasgow, Glasgow G12 8QQ, UK
| | - Nita G Forouhi
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Francesco Gianfagna
- Department of Clinical and Experimental Medicine, Research Centre in Epidemiology and Preventive Medicine, University of Insubria, 21100 Varese, Italy. Department of Epidemiology and Prevention, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy
| | | | - Sara Grioni
- Epidemiology and Prevention Unit, 20133 Milan, Italy
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Jan-Håkan Jansson
- Research Unit, 931 41 Skellefteå, Sweden. Department of Public Health & Clinical Medicine, Umeå University, 901 85 Umeå, Sweden
| | - Marit E Jørgensen
- Steno Diabetes Center, 2820 Gentofte, Denmark. National Institute of Public Health, Southern Denmark University, DK-1353 Odense, Denmark
| | - J Wouter Jukema
- Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Rudolf Kaaks
- German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
| | - Frank Kee
- UK Clinical Research Collaboration (UKCRC) Centre of Excellence for Public Health, Queen's University Belfast, Northern Ireland, Belfast BT12 6BJ, UK
| | - Nicola D Kerrison
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | | | - Jukka Kontto
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | | | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Kari Kuulasmaa
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland. Kuopio University Hospital, FL 70029 Kuopio, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region, DK-2600 Copenhagen, Denmark. Department of Clinical Experimental Research, Rigshospitalet, 2100 Glostrup, Denmark. Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Chunyu Liu
- Framingham Heart Study, Population Sciences Branch, NHLBI/National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Gaëlle Marenne
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-7264, USA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK. Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Kenneth Muir
- Centre for Epidemiology, Institute of Population Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK. University of Warwick, Coventry CV4 7AL, UK
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany. Department of Medicine I, Ludwig Maximilians University Munich, 80336 Munich, Germany. DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Carmen Navarro
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain. Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia 30008, Spain
| | - Sune F Nielsen
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, University of Copenhagen, 2730 Copenhagen, Denmark
| | | | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, University of Copenhagen, 2730 Copenhagen, Denmark
| | | | - Domenico Palli
- Cancer Research and Prevention Institute (ISPO), 50141 Florence, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Gina M Peloso
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA. Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA. Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Markus Perola
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland. Institute of Molecular Medicine Finland (FIMM), University of Helsinki, FI-00014 Helsinki, Finland
| | - Annette Peters
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany. Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Christopher J Poole
- University of Warwick, Coventry CV4 7AL, UK. Department of Medical Oncology, Arden Cancer Centre, University Hospital Coventry and Warwickshire, West Midlands CV2 2DX, UK
| | - J Ramón Quirós
- Public Health Directorate, 33006 Oviedo, Asturias, Spain
| | | | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Citta' della Salute e della Scienza Hospital, University of Turin, 10126 Torino, Italy. Center for Cancer Prevention (CPO), 10126 Torino, Italy. Human Genetics Foundation, 10126 Torino, Italy
| | - Veikko Salomaa
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - María-José Sánchez
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain. Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria ibs.GRANADA. Hospitales Universitarios de Granada/Universidad de Granada, Granada 18012, Spain
| | | | - Stephen J Sharp
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Rebecca Sims
- Institute of Psychological Medicine and Clinical Neuroscience, MRC Centre, Cardiff University, Cardiff CF24 4HQ, UK
| | - Nadia Slimani
- International Agency for Research on Cancer, 69372 Lyon, France
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109-2029, USA
| | - Deborah J Thompson
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Stella Trompet
- Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic-M.P. Arezzo" Hospital, ASP Ragusa, 97100 Ragusa, Italy
| | - Daphne L van der A
- National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, Netherlands
| | | | - Jarmo Virtamo
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Klaudia Walter
- The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Jean E Abraham
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jennifer L Aponte
- Genetics, PCPS, GlaxoSmithKline, Research Triangle Park, NC 27709, USA
| | - Adam S Butterworth
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK. Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Rosalind A Eeles
- The Institute of Cancer Research, London SM2 5NG, UK. Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey SW3 6JJ, UK
| | - Jeanette Erdmann
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 Malmö, Sweden. Department of Public Health & Clinical Medicine, Umeå University, 901 85 Umeå, Sweden. Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Joanna M M Howson
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK
| | - Torben Jørgensen
- Research Centre for Prevention and Health, DK-2600 Capital Region, Denmark. Department of Public Health, Institute of Health Science, University of Copenhagen, 1014 Copenhagen, Denmark. Faculty of Medicine, Aalborg University, 9220 Aalborg, Denmark
| | - Jaspal Kooner
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK. Imperial College Healthcare NHS Trust, London W2 1NY, UK. Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Markku Laakso
- Department of Medicine, University of Kuopio, FI-70211 Kuopio, Finland
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK. Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford, UK
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455-0381, USA
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elio Riboli
- School of Public Health, Imperial College London, London W2 1PG, UK
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles Medical Center, Torrance, CA 90502, USA
| | - Danish Saleheen
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK. National Institute for Health Research, Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany. Deutsches Herzzentrum München, Technische Universität München, 80636 Munich, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, BH10-462, Internal Medicine, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland
| | - Stephen O'Rahilly
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge CB2 0QQ, UK. MRC Metabolic Diseases Unit, Cambridge CB2 0QQ, UK. National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John Danesh
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge CB1 8RN, UK. The Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sekar Kathiresan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA. Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA. Cardiology Division, Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - James B Meigs
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Margaret G Ehm
- Genetics, PCPS, GlaxoSmithKline, Research Triangle Park, NC 27709, USA
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
| | | |
Collapse
|
441
|
Jenkinson CP, Göring HH, Arya R, Blangero J, Duggirala R, DeFronzo RA. Transcriptomics in type 2 diabetes: Bridging the gap between genotype and phenotype. GENOMICS DATA 2016; 8:25-36. [PMID: 27114903 PMCID: PMC4832048 DOI: 10.1016/j.gdata.2015.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/19/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is a common, multifactorial disease that is influenced by genetic and environmental factors and their interactions. However, common variants identified by genome wide association studies (GWAS) explain only about 10% of the total trait variance for T2D and less than 5% of the variance for obesity, indicating that a large proportion of heritability is still unexplained. The transcriptomic approach described here uses quantitative gene expression and disease-related physiological data (deep phenotyping) to measure the direct correlation between the expression of specific genes and physiological traits. Transcriptomic analysis bridges the gulf between GWAS and physiological studies. Recent GWAS studies have utilized very large population samples, numbering in the tens of thousands (or even hundreds of thousands) of individuals, yet establishing causal functional relationships between strongly associated genetic variants and disease remains elusive. In light of the findings described below, it is appropriate to consider how and why transcriptomic approaches in small samples might be capable of identifying complex disease-related genes which are not apparent using GWAS in large samples.
Collapse
Affiliation(s)
- Christopher P. Jenkinson
- South Texas Diabetes and Obesity Institute (STDOI), University of Texas Rio Grande Valley (UTRGV), TX, USA
| | - Harald H.H. Göring
- South Texas Diabetes and Obesity Institute (STDOI), University of Texas Rio Grande Valley (UTRGV), TX, USA
| | - Rector Arya
- South Texas Diabetes and Obesity Institute (STDOI), University of Texas Rio Grande Valley (UTRGV), TX, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute (STDOI), University of Texas Rio Grande Valley (UTRGV), TX, USA
| | - Ravindranath Duggirala
- South Texas Diabetes and Obesity Institute (STDOI), University of Texas Rio Grande Valley (UTRGV), TX, USA
| | - Ralph A. DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
442
|
Sung YJ, Winkler TW, Manning AK, Aschard H, Gudnason V, Harris TB, Smith AV, Boerwinkle E, Brown MR, Morrison AC, Fornage M, Lin LA, Richard M, Bartz TM, Psaty BM, Hayward C, Polasek O, Marten J, Rudan I, Feitosa MF, Kraja AT, Province MA, Deng X, Fisher VA, Zhou Y, Bielak LF, Smith J, Huffman JE, Padmanabhan S, Smith BH, Ding J, Liu Y, Lohman K, Bouchard C, Rankinen T, Rice TK, Arnett D, Schwander K, Guo X, Palmas W, Rotter JI, Alfred T, Bottinger EP, Loos RJF, Amin N, Franco OH, van Duijn CM, Vojinovic D, Chasman DI, Ridker PM, Rose LM, Kardia S, Zhu X, Rice K, Borecki IB, Rao DC, Gauderman WJ, Cupples LA. An Empirical Comparison of Joint and Stratified Frameworks for Studying G × E Interactions: Systolic Blood Pressure and Smoking in the CHARGE Gene-Lifestyle Interactions Working Group. Genet Epidemiol 2016; 40:404-15. [PMID: 27230302 DOI: 10.1002/gepi.21978] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/08/2016] [Accepted: 04/04/2016] [Indexed: 01/11/2023]
Abstract
Studying gene-environment (G × E) interactions is important, as they extend our knowledge of the genetic architecture of complex traits and may help to identify novel variants not detected via analysis of main effects alone. The main statistical framework for studying G × E interactions uses a single regression model that includes both the genetic main and G × E interaction effects (the "joint" framework). The alternative "stratified" framework combines results from genetic main-effect analyses carried out separately within the exposed and unexposed groups. Although there have been several investigations using theory and simulation, an empirical comparison of the two frameworks is lacking. Here, we compare the two frameworks using results from genome-wide association studies of systolic blood pressure for 3.2 million low frequency and 6.5 million common variants across 20 cohorts of European ancestry, comprising 79,731 individuals. Our cohorts have sample sizes ranging from 456 to 22,983 and include both family-based and population-based samples. In cohort-specific analyses, the two frameworks provided similar inference for population-based cohorts. The agreement was reduced for family-based cohorts. In meta-analyses, agreement between the two frameworks was less than that observed in cohort-specific analyses, despite the increased sample size. In meta-analyses, agreement depended on (1) the minor allele frequency, (2) inclusion of family-based cohorts in meta-analysis, and (3) filtering scheme. The stratified framework appears to approximate the joint framework well only for common variants in population-based cohorts. We conclude that the joint framework is the preferred approach and should be used to control false positives when dealing with low-frequency variants and/or family-based cohorts.
Collapse
Affiliation(s)
- Yun Ju Sung
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Thomas W Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Alisa K Manning
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America.,Center for Human Genetics Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America.,Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Li-An Lin
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Traci M Bartz
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia.,Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jonathan Marten
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Igor Rudan
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Virginia A Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Jennifer Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Jennifer E Huffman
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Blair H Smith
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Kurt Lohman
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Los Angeles, United States of America
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Los Angeles, United States of America
| | - Treva K Rice
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Donna Arnett
- Department of Epidemiology, University of Alabama-Birmingham, Birmingham, Alabama, United States of America
| | - Karen Schwander
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Xiuqing Guo
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Jerome I Rotter
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Tamuno Alfred
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America.,The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Oscar H Franco
- Cardiovascular Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Dina Vojinovic
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Daniel I Chasman
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Paul M Ridker
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Lynda M Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Sharon Kardia
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kenneth Rice
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - W James Gauderman
- Department of Preventive Medicine, University of Southern California, Los Angeles, California, United States of America
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America.,Framingham Heart Study, Framingham, Massachusetts, United States of America
| |
Collapse
|
443
|
Arda HE, Li L, Tsai J, Torre EA, Rosli Y, Peiris H, Spitale RC, Dai C, Gu X, Qu K, Wang P, Wang J, Grompe M, Scharfmann R, Snyder MS, Bottino R, Powers AC, Chang HY, Kim SK. Age-Dependent Pancreatic Gene Regulation Reveals Mechanisms Governing Human β Cell Function. Cell Metab 2016; 23:909-20. [PMID: 27133132 PMCID: PMC4864151 DOI: 10.1016/j.cmet.2016.04.002] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/03/2015] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
Intensive efforts are focused on identifying regulators of human pancreatic islet cell growth and maturation to accelerate development of therapies for diabetes. After birth, islet cell growth and function are dynamically regulated; however, establishing these age-dependent changes in humans has been challenging. Here, we describe a multimodal strategy for isolating pancreatic endocrine and exocrine cells from children and adults to identify age-dependent gene expression and chromatin changes on a genomic scale. These profiles revealed distinct proliferative and functional states of islet α cells or β cells and histone modifications underlying age-dependent gene expression changes. Expression of SIX2 and SIX3, transcription factors without prior known functions in the pancreas and linked to fasting hyperglycemia risk, increased with age specifically in human islet β cells. SIX2 and SIX3 were sufficient to enhance insulin content or secretion in immature β cells. Our work provides a unique resource to study human-specific regulators of islet cell maturation and function.
Collapse
Affiliation(s)
- H Efsun Arda
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lingyu Li
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer Tsai
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Eduardo A Torre
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yenny Rosli
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heshan Peiris
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert C Spitale
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chunhua Dai
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kun Qu
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pei Wang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jing Wang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Markus Grompe
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Michael S Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA
| | - Alvin C Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Howard Y Chang
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine (Oncology Division), Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
444
|
White MJ, Risse-Adams O, Goddard P, Contreras MG, Adams J, Hu D, Eng C, Oh SS, Davis A, Meade K, Brigino-Buenaventura E, LeNoir MA, Bibbins-Domingo K, Pino-Yanes M, Burchard EG. Novel genetic risk factors for asthma in African American children: Precision Medicine and the SAGE II Study. Immunogenetics 2016; 68:391-400. [PMID: 27142222 DOI: 10.1007/s00251-016-0914-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/25/2016] [Indexed: 01/06/2023]
Abstract
Asthma, an inflammatory disorder of the airways, is the most common chronic disease of children worldwide. There are significant racial/ethnic disparities in asthma prevalence, morbidity, and mortality among US children. This trend is mirrored in obesity, which may share genetic and environmental risk factors with asthma. The majority of asthma biomedical research has been performed in populations of European decent. We sought to identify genetic risk factors for asthma in African American children. We also assessed the generalizability of genetic variants associated with asthma in European and Asian populations to African American children. Our study population consisted of 1227 (812 asthma cases, 415 controls) African American children with genome-wide single nucleotide polymorphism (SNP) data. Logistic regression was used to identify associations between SNP genotype and asthma status. We identified a novel variant in the PTCHD3 gene that is significantly associated with asthma (rs660498, p = 2.2 × 10(-7)) independent of obesity status. Approximately 5 % of previously reported asthma genetic associations identified in European populations replicated in African Americans. Our identification of novel variants associated with asthma in African American children, coupled with our inability to replicate the majority of findings reported in European Americans, underscores the necessity for including diverse populations in biomedical studies of asthma.
Collapse
Affiliation(s)
- Marquitta J White
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA.
| | - O Risse-Adams
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- Lowell Science Research Program, Lowell High School, San Francisco, CA, USA
| | - P Goddard
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - M G Contreras
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- SF BUILD, San Francisco State University, San Francisco, CA, USA
| | - J Adams
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - D Hu
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - C Eng
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - S S Oh
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - A Davis
- Children's Hospital and Research Center Oakland, Oakland, CA, USA
| | - K Meade
- Children's Hospital and Research Center Oakland, Oakland, CA, USA
| | - E Brigino-Buenaventura
- Department of Allergy and Immunology, Kaiser Permanente Vallejo Medical Center, Vallejo, CA, USA
| | | | - K Bibbins-Domingo
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - M Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - E G Burchard
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
445
|
Yang RY, Xue H, Yu L, Velayos-Baeza A, Monaco AP, Liu FT. Identification of VPS13C as a Galectin-12-Binding Protein That Regulates Galectin-12 Protein Stability and Adipogenesis. PLoS One 2016; 11:e0153534. [PMID: 27073999 PMCID: PMC4830523 DOI: 10.1371/journal.pone.0153534] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
Galectin-12, a member of the galectin family of β-galactoside-binding animal lectins, is preferentially expressed in adipocytes and required for adipocyte differentiation in vitro. This protein was recently found to regulate lipolysis, whole body adiposity, and glucose homeostasis in vivo. Here we identify VPS13C, a member of the VPS13 family of vacuolar protein sorting-associated proteins highly conserved throughout eukaryotic evolution, as a major galectin-12-binding protein. VPS13C is upregulated during adipocyte differentiation, and is required for galectin-12 protein stability. Knockdown of Vps13c markedly reduces the steady-state levels of galectin-12 by promoting its degradation through primarily the lysosomal pathway, and impairs adipocyte differentiation. Our studies also suggest that VPS13C may have a broader role in protein quality control. The regulation of galectin-12 stability by VPS13C could potentially be exploited for therapeutic intervention of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Ri-Yao Yang
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | - Huiting Xue
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- School of Life Sciences, Northeast Normal University, Changchun, 130024, People’s Republic of China
| | - Lan Yu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | | | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, OX3 7BN, Oxford, United Kingdom
| | - Fu-Tong Liu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| |
Collapse
|
446
|
Szendroedi J, Saxena A, Weber KS, Strassburger K, Herder C, Burkart V, Nowotny B, Icks A, Kuss O, Ziegler D, Al-Hasani H, Müssig K, Roden M. Cohort profile: the German Diabetes Study (GDS). Cardiovasc Diabetol 2016; 15:59. [PMID: 27053136 PMCID: PMC4823856 DOI: 10.1186/s12933-016-0374-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/24/2016] [Indexed: 12/16/2022] Open
Abstract
Background The German Diabetes Study (GDS) is a prospective longitudinal cohort study describing the impact of subphenotypes on the course of the disease. GDS aims at identifying prognostic factors and mechanisms underlying the development of related comorbidities. Study design and methods The study comprises intensive phenotyping within 12 months after clinical diagnosis, at 5-year intervals for 20 years and annual telephone interviews in between. Dynamic tests, including glucagon, mixed meal, intravenous glucose tolerance and hyperinsulinemic clamp tests, serve to assess beta-cell function and tissue-specific insulin sensitivity. Magnetic resonance imaging and multinuclei spectroscopy allow quantifying whole-body fat distribution, tissue-specific lipid deposition and energy metabolism. Comprehensive analyses of microvascular (nerve, eye, kidney) and macrovascular (endothelial, cardiorespiratory) morphology and function enable identification and monitoring of comorbidities. The GDS biobank stores specimens from blood, stool, skeletal muscle, subcutaneous adipose tissue and skin for future analyses including multiomics, expression profiles and histology. Repeated questionnaires on socioeconomic conditions, patient-reported outcomes as quality of life, health-related behavior as physical activity and nutritional habits are a specific asset of GDS. This study will recruit 3000 patients and a group of humans without familiy history of diabetes. 237 type 1 and 456 type 2 diabetes patients have been already included. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0374-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Szendroedi
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Aaruni Saxena
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Katharina S Weber
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Bettina Nowotny
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Andrea Icks
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,Public Health Unit, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Oliver Kuss
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Hadi Al-Hasani
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry German Diabetes Center, Leibniz Institute for Diabetes Research, Düsseldorf, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, Leibniz Institute for Diabetes Research, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany. .,German Center for Diabetes Research (DZD), München-Neuherberg, Germany. .,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| | | |
Collapse
|
447
|
Risk Alleles in/near ADCY5, ADRA2A, CDKAL1, CDKN2A/B, GRB10, and TCF7L2 Elevate Plasma Glucose Levels at Birth and in Early Childhood: Results from the FAMILY Study. PLoS One 2016; 11:e0152107. [PMID: 27049325 PMCID: PMC4822946 DOI: 10.1371/journal.pone.0152107] [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: 12/03/2015] [Accepted: 02/20/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Metabolic abnormalities that lead to type 2 diabetes mellitus begin in early childhood. OBJECTIVES We investigate whether common genetic variants identified in adults have an effect on glucose in early life. METHODS 610 newborns, 463 mothers, and 366 fathers were included in the present study. Plasma glucose and anthropometric characteristics were collected at birth, 3, and 5 years. After quality assessment, 37 SNPs, which have demonstrated an association with fasting plasma glucose at the genome-wide threshold in adults, were studied. Quantitative trait disequilibrium tests and mixed-effects regressions were conducted to estimate an effect of the SNPs on glucose. RESULTS Risk alleles for 6 loci increased glucose levels from birth to 5 years of age (ADCY5, ADRA2A, CDKAL1, CDKN2A/B, GRB10, and TCF7L2, 4.85x10-3 ≤ P ≤ 4.60x10-2). Together, these 6 SNPs increase glucose by 0.05 mmol/L for each risk allele in a genotype score (P = 6.33x10-5). None of the associations described in the present study have been reported previously in early childhood. CONCLUSION Our data support the notion that a subset of loci contributing to plasma glucose variation in adults has an effect at birth and in early life.
Collapse
|
448
|
Dunn EC, Wiste A, Radmanesh F, Almli LM, Gogarten SM, Sofer T, Faul JD, Kardia SL, Smith JA, Weir DR, Zhao W, Soare TW, Mirza SS, Hek K, Tiemeier HW, Goveas JS, Sarto GE, Snively BM, Cornelis M, Koenen KC, Kraft P, Purcell S, Ressler KJ, Rosand J, Wassertheil-Smoller S, Smoller JW. GENOME-WIDE ASSOCIATION STUDY (GWAS) AND GENOME-WIDE BY ENVIRONMENT INTERACTION STUDY (GWEIS) OF DEPRESSIVE SYMPTOMS IN AFRICAN AMERICAN AND HISPANIC/LATINA WOMEN. Depress Anxiety 2016; 33:265-80. [PMID: 27038408 PMCID: PMC4826276 DOI: 10.1002/da.22484] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have made little progress in identifying variants linked to depression. We hypothesized that examining depressive symptoms and considering gene-environment interaction (GxE) might improve efficiency for gene discovery. We therefore conducted a GWAS and genome-wide by environment interaction study (GWEIS) of depressive symptoms. METHODS Using data from the SHARe cohort of the Women's Health Initiative, comprising African Americans (n = 7,179) and Hispanics/Latinas (n = 3,138), we examined genetic main effects and GxE with stressful life events and social support. We also conducted a heritability analysis using genome-wide complex trait analysis (GCTA). Replication was attempted in four independent cohorts. RESULTS No SNPs achieved genome-wide significance for main effects in either discovery sample. The top signals in African Americans were rs73531535 (located 20 kb from GPR139, P = 5.75 × 10(-8) ) and rs75407252 (intronic to CACNA2D3, P = 6.99 × 10(-7) ). In Hispanics/Latinas, the top signals were rs2532087 (located 27 kb from CD38, P = 2.44 × 10(-7) ) and rs4542757 (intronic to DCC, P = 7.31 × 10(-7) ). In the GEWIS with stressful life events, one interaction signal was genome-wide significant in African Americans (rs4652467; P = 4.10 × 10(-10) ; located 14 kb from CEP350). This interaction was not observed in a smaller replication cohort. Although heritability estimates for depressive symptoms and stressful life events were each less than 10%, they were strongly genetically correlated (rG = 0.95), suggesting that common variation underlying self-reported depressive symptoms and stressful life event exposure, though modest on their own, were highly overlapping in this sample. CONCLUSIONS Our results underscore the need for larger samples, more GEWIS, and greater investigation into genetic and environmental determinants of depressive symptoms in minorities.
Collapse
Affiliation(s)
- Erin C. Dunn
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| | - Anna Wiste
- Center for Experimental Drugs and Diagnostics, Department of Psychiatry, Massachusetts General Hospital
| | - Farid Radmanesh
- Center for Human Genetic Research, Massachusetts General Hospital
- Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT
| | - Lynn M. Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | | | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Jessica D. Faul
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | | | - Jennifer A. Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - David R. Weir
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Thomas W. Soare
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| | - Saira S. Mirza
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Karin Hek
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Henning W. Tiemeier
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joseph S. Goveas
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gloria E. Sarto
- Center for Women's Health and Health Disparities Research, Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Beverly M. Snively
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Marilyn Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Karestan C. Koenen
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
| | - Shaun Purcell
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kerry J. Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Jonathan Rosand
- Center for Human Genetic Research, Massachusetts General Hospital
- Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York
| | - Jordan W. Smoller
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| |
Collapse
|
449
|
Fan Q, Verhoeven VJM, Wojciechowski R, Barathi VA, Hysi PG, Guggenheim JA, Höhn R, Vitart V, Khawaja AP, Yamashiro K, Hosseini SM, Lehtimäki T, Lu Y, Haller T, Xie J, Delcourt C, Pirastu M, Wedenoja J, Gharahkhani P, Venturini C, Miyake M, Hewitt AW, Guo X, Mazur J, Huffman JE, Williams KM, Polasek O, Campbell H, Rudan I, Vatavuk Z, Wilson JF, Joshi PK, McMahon G, St Pourcain B, Evans DM, Simpson CL, Schwantes-An TH, Igo RP, Mirshahi A, Cougnard-Gregoire A, Bellenguez C, Blettner M, Raitakari O, Kähönen M, Seppala I, Zeller T, Meitinger T, Consortium for Refractive Error and Myopia (CREAM), Ried JS, Gieger C, Portas L, van Leeuwen EM, Amin N, Uitterlinden AG, Rivadeneira F, Hofman A, Vingerling JR, Wang YX, Wang X, Tai-Hui Boh E, Ikram MK, Sabanayagam C, Gupta P, Tan V, Zhou L, Ho CEH, Lim W, Beuerman RW, Siantar R, Tai ES, Vithana E, Mihailov E, Khor CC, Hayward C, Luben RN, Foster PJ, Klein BEK, Klein R, Wong HS, Mitchell P, Metspalu A, Aung T, Young TL, He M, Pärssinen O, van Duijn CM, Jin Wang J, Williams C, Jonas JB, Teo YY, Mackey DA, Oexle K, Yoshimura N, Paterson AD, Pfeiffer N, Wong TY, Baird PN, Stambolian D, Wilson JEB, Cheng CY, et alFan Q, Verhoeven VJM, Wojciechowski R, Barathi VA, Hysi PG, Guggenheim JA, Höhn R, Vitart V, Khawaja AP, Yamashiro K, Hosseini SM, Lehtimäki T, Lu Y, Haller T, Xie J, Delcourt C, Pirastu M, Wedenoja J, Gharahkhani P, Venturini C, Miyake M, Hewitt AW, Guo X, Mazur J, Huffman JE, Williams KM, Polasek O, Campbell H, Rudan I, Vatavuk Z, Wilson JF, Joshi PK, McMahon G, St Pourcain B, Evans DM, Simpson CL, Schwantes-An TH, Igo RP, Mirshahi A, Cougnard-Gregoire A, Bellenguez C, Blettner M, Raitakari O, Kähönen M, Seppala I, Zeller T, Meitinger T, Consortium for Refractive Error and Myopia (CREAM), Ried JS, Gieger C, Portas L, van Leeuwen EM, Amin N, Uitterlinden AG, Rivadeneira F, Hofman A, Vingerling JR, Wang YX, Wang X, Tai-Hui Boh E, Ikram MK, Sabanayagam C, Gupta P, Tan V, Zhou L, Ho CEH, Lim W, Beuerman RW, Siantar R, Tai ES, Vithana E, Mihailov E, Khor CC, Hayward C, Luben RN, Foster PJ, Klein BEK, Klein R, Wong HS, Mitchell P, Metspalu A, Aung T, Young TL, He M, Pärssinen O, van Duijn CM, Jin Wang J, Williams C, Jonas JB, Teo YY, Mackey DA, Oexle K, Yoshimura N, Paterson AD, Pfeiffer N, Wong TY, Baird PN, Stambolian D, Wilson JEB, Cheng CY, Hammond CJ, Klaver CCW, Saw SM, Rahi JS, Korobelnik JF, Kemp JP, Timpson NJ, Smith GD, Craig JE, Burdon KP, Fogarty RD, Iyengar SK, Chew E, Janmahasatian S, Martin NG, MacGregor S, Xu L, Schache M, Nangia V, Panda-Jonas S, Wright AF, Fondran JR, Lass JH, Feng S, Zhao JH, Khaw KT, Wareham NJ, Rantanen T, Kaprio J, Pang CP, Chen LJ, Tam PO, Jhanji V, Young AL, Döring A, Raffel LJ, Cotch MF, Li X, Yip SP, Yap MK, Biino G, Vaccargiu S, Fossarello M, Fleck B, Yazar S, Tideman JWL, Tedja M, Deangelis MM, Morrison M, Farrer L, Zhou X, Chen W, Mizuki N, Meguro A, Mäkelä KM. Meta-analysis of gene-environment-wide association scans accounting for education level identifies additional loci for refractive error. Nat Commun 2016; 7:11008. [PMID: 27020472 PMCID: PMC4820539 DOI: 10.1038/ncomms11008] [Show More Authors] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/10/2016] [Indexed: 02/07/2023] Open
Abstract
Myopia is the most common human eye disorder and it results from complex genetic and environmental causes. The rapidly increasing prevalence of myopia poses a major public health challenge. Here, the CREAM consortium performs a joint meta-analysis to test single-nucleotide polymorphism (SNP) main effects and SNP × education interaction effects on refractive error in 40,036 adults from 25 studies of European ancestry and 10,315 adults from 9 studies of Asian ancestry. In European ancestry individuals, we identify six novel loci (FAM150B-ACP1, LINC00340, FBN1, DIS3L-MAP2K1, ARID2-SNAT1 and SLC14A2) associated with refractive error. In Asian populations, three genome-wide significant loci AREG, GABRR1 and PDE10A also exhibit strong interactions with education (P<8.5 × 10(-5)), whereas the interactions are less evident in Europeans. The discovery of these loci represents an important advance in understanding how gene and environment interactions contribute to the heterogeneity of myopia.
Collapse
Affiliation(s)
- Qiao Fan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Virginie J. M. Verhoeven
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Robert Wojciechowski
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 20205, USA
| | - Veluchamy A. Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
| | - Jeremy A. Guggenheim
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - René Höhn
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
- Department of Ophthalmology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Anthony P. Khawaja
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - S Mohsen Hosseini
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Yi Lu
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Jing Xie
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Cécile Delcourt
- Université de Bordeaux, ISPED (Institut de Santé Publique d'Épidémiologie et de Développement), Bordeaux 33000, France
- INSERM, U1219-Bordeaux Population Health Research Center, Bordeaux 33000, France
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Juho Wedenoja
- Department of Public Health, University of Helsinki, Helsinki 00014, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Puya Gharahkhani
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- UCL Institute of Ophthalmology, London SE1 7EH, UK
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Alex W. Hewitt
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Xiaobo Guo
- Department of Statistical Science, School of Mathematics and Computational Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Johanna Mazur
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131 Mainz, Germany
| | - Jenifer E. Huffman
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Katie M. Williams
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- Department of Ophthalmology, King's College London, London SE1 7EH, UK
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Harry Campbell
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Igor Rudan
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Zoran Vatavuk
- Department of Ophthalmology, Sisters of Mercy University Hospital, Zagreb 10000, Croatia
| | - James F. Wilson
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Peter K. Joshi
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - George McMahon
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
- Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
| | - David M. Evans
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - Claire L. Simpson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Tae-Hwi Schwantes-An
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Robert P. Igo
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Alireza Mirshahi
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
- Dardenne Eye Hospital, Bonn-Bad Godesberg, 53177 Bonn, Germany
| | - Audrey Cougnard-Gregoire
- Université de Bordeaux, ISPED (Institut de Santé Publique d'Épidémiologie et de Développement), Bordeaux 33000, France
- INSERM, U1219-Bordeaux Population Health Research Center, Bordeaux 33000, France
| | - Céline Bellenguez
- Inserm, U1167, Lille 59000, France
- Univ. Lille, U1167, Lille 59000, France
- Université Lille 2, Lille 59000, France
| | - Maria Blettner
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131 Mainz, Germany
| | - Olli Raitakari
- Research Centre of Applied and Preventive Medicine, University of Turku, Turku 20520, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Ilkka Seppala
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | | | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Laura Portas
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | | | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
| | - Xu Wang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Eileen Tai-Hui Boh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - M. Kamran Ikram
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Preeti Gupta
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Vincent Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Lei Zhou
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Candice E. H. Ho
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Wan'e Lim
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Roger W. Beuerman
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Rosalynn Siantar
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - E-Shyong Tai
- Duke-NUS Medical School, Singapore 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Department of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Eranga Vithana
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Chiea-Chuen Khor
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Robert N. Luben
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Paul J. Foster
- Division of Genetics and Epidemiology, UCL Institute of Ophthalmology, London EC1V 9EL, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 2PD, UK
| | - Barbara E. K. Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53726, USA
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53726, USA
| | - Hoi-Suen Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Paul Mitchell
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Terri L. Young
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, USA
| | - Mingguang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Olavi Pärssinen
- Department of Ophthalmology, Central Hospital of Central Finland, Jyväskylä 40620, Finland
- Gerontology Research Center and Department of Health Sciences, University of Jyväskylä, Jyväskylä 40014, Finland
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Jie Jin Wang
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Cathy Williams
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Jost B. Jonas
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
- Medical Faculty Mannheim, Department of Ophthalmology, Ruprecht-Karls-University Heidelberg, 69115 Mannheim, Germany
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore 117546, Singapore
| | - David A. Mackey
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Konrad Oexle
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Nagahisa Yoshimura
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Andrew D. Paterson
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Paul N. Baird
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Joan E. Bailey Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Christopher J. Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- Department of Ophthalmology, King's College London, London SE1 7EH, UK
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Seang-Mei Saw
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Jugnoo S. Rahi
- Medical Research Council Centre of Epidemiology for Child Health, Institute of Child Health, University College London, London WC1E 6BT, UK
- Institute of Ophthalmology, Moorfields Eye Hospital, London EC1V 2PD, UK
- Ulverscroft Vision Research Group, University College London, London WC1E 6BT, UK
| | - Jean-François Korobelnik
- Université de Bordeaux, 33400 Talence, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), ISPED (Institut de Santé Publique d'épidémiologie et de Développement), Centre INSERM U897-Epidemiologie-Biostatistique, 33076 Bordeaux, France
| | - John P. Kemp
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Kathryn P. Burdon
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Rhys D. Fogarty
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Sudha K. Iyengar
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio 44106, USA
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Emily Chew
- National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sarayut Janmahasatian
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
| | - Nicholas G. Martin
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Stuart MacGregor
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Liang Xu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
| | - Maria Schache
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Vinay Nangia
- Suraj Eye Institute, Nagpur, Maharashtra 440001, India
| | | | - Alan F. Wright
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Jeremy R. Fondran
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
| | - Jonathan H. Lass
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio 44106, USA
| | - Sheng Feng
- Department of Pediatric Ophthalmology, Duke Eye Center For Human Genetics, Durham, North Carolina 27710, USA
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Sciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Nick J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Taina Rantanen
- Gerontology Research Center, University of Jyväskylä, Jyväskylä Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki 00014, Finland
- Institute for Molecular Medicine, University of Helsinki, Helsinki 00014, Finland
- Department of Mental Health and Alcohol Abuse Services, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Pancy O. Tam
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Vishal Jhanji
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Alvin L. Young
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - 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
| | - Leslie J. Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Mary-Frances Cotch
- Division of Epidemiology and Clinical Applications, National Eye Institute, Bethesda, Maryland 20892, USA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Los Angeles, California 90502, USA
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Maurice K.H. Yap
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Simona Vaccargiu
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Maurizio Fossarello
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Brian Fleck
- Princess Alexandra Eye Pavilion, Edinburgh EH3 9HA, UK
| | - Seyhan Yazar
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Jan Willem L. Tideman
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Milly Tedja
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Margaret M. Deangelis
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, Utah 84132, USA
| | - Margaux Morrison
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah 84132, USA
| | - Lindsay Farrer
- Departments of Medicine (Biomedical Genetics), Ophthalmology, Neurology, Epidemiology and Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts 02118, USA
| | - Xiangtian Zhou
- School of ophthalmology and optometry, Wenzhou Medical University, Wenzhou 325035, China
| | - Wei Chen
- School of ophthalmology and optometry, Wenzhou Medical University, Wenzhou 325035, China
| | - Nobuhisa Mizuki
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0027, Japan
| | - Akira Meguro
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0027, Japan
| | - Kari Matti Mäkelä
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33014, Finland
| |
Collapse
|
450
|
Fareed M, Afzal M. Increased cardiovascular risks associated with familial inbreeding: a population-based study of adolescent cohort. Ann Epidemiol 2016; 26:283-92. [PMID: 27084548 DOI: 10.1016/j.annepidem.2016.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 01/03/2023]
Abstract
PURPOSE Cardiovascular diseases are the leading cause of mortality and morbidity among humans worldwide. We aimed to estimate the effect of familial inbreeding on cardiovascular risks. METHODS The study was conducted during April 2014 through June 2014, and a total of 587 adolescent subjects (male = 270, female = 317; 11-18 years of age) were recruited from five Muslim populations viz., Gujjar and Bakarwal (n = 130), Mughal (n = 111), Malik (n = 114), Syed (n = 108), and Khan (n = 124). Wright's path relationship method was used for calculating the coefficient of inbreeding (F). Anthropometric and physiological parameters were estimated using standard methods. RESULTS We observed higher mean values for major physiological traits among the inbred subjects in comparison with the non-inbred groups of five different populations. Our study suggests that inbreeding and sex are the key factors affecting cardiovascular profile. Multivariate analysis of covariance revealed inbreeding as a major source of variation for cardiovascular risks, dominating over other factors causing greater variability in the physiological traits. The magnitude of cardiovascular risks shows an increase with the increase in the values of coefficient of inbreeding (i.e., from F = 0.00 to F = 0.125). The abnormal levels of systolic blood pressure (SBP; range 140-159 mm Hg) and fasting blood glucose (FBG; range 101-126 mg per dL) show persuasive increase with an upsurge in the homozygosity level (i.e., coefficient of inbreeding). CONCLUSIONS Our comprehensive assessment presents the deleterious consequence of inbreeding on cardiovascular profile. This study can be used as fact-sheet for framing the heath policies and hence can play a vital role in genetic counseling strategies for transforming the public opinion regarding the practice of consanguinity and its associated risks.
Collapse
Affiliation(s)
- Mohd Fareed
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| |
Collapse
|