151
|
Beysel S, Eyerci N, Pinarli FA, Kizilgul M, Ozcelik O, Caliskan M, Cakal E. HNF1A gene p.I27L is associated with early-onset, maturity-onset diabetes of the young-like diabetes in Turkey. BMC Endocr Disord 2019; 19:51. [PMID: 31109344 PMCID: PMC6528345 DOI: 10.1186/s12902-019-0375-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The molecular basis of the Turkish population with suspected maturity-onset diabetes of the young (MODY) has not been identified. This is the first study to investigate the association between HNF1A-gene single-nucleotide polymorphisms (SNPs) and having early-onset, MODY-like diabetes mellitus in the Turkish population. METHODS All diabetic patients (N = 565) who presented to our clinic between 2012 and 2015 with a clinical suspicion of MODY were included in the study. Analysis of HNF1A, HNFB, HNF4A, GCK gene mutations was performed using real-time polymerase chain reaction sequencing. After genetic analysis, diabetics (n = 46) with HNF1A, HNF1B, HNF4A, GCK gene mutations (diagnosed as MODY) and diabetics (n = 30) with HNF1B, HNF4A, GCK gene SNPs were excluded. Patients with early-onset, MODY-like diabetes (n = 486) and non-diabetic controls (n = 263) were included. Genetic analyses for the HNF1A gene p.S487 N (rs2464196), p.A98V (rs1800574) and p.I27L (rs1169288) SNPs were performed using Sanger-based DNA sequencing among the control group. RESULTS p.S487 N and p.A98V was similar between the diabetics and controls in dominant and recessive models with no association (each, p > 0.05). p.I27L GT/TT carriers (GT/TT vs. GG, OR = 1.68, 95% CI: [1. 21-2.13]; p = 0.035) and p.I27L TT carriers had increased risk of having MODY-like diabetes (GT/GG vs. TT, OR = 1.56, 95% CI: [1. 14-2.57]; p = 0.048). Family inheritance of diabetes was significantly more common in patients with the p.I27L TT genotype. The p.I27L SNP was modestly associated with having diabetes after adjusting for body mass index and age (β = 1.45, 95% CI: [1. 2-4.2]; p = 0.036). CONCLUSIONS The HNF1A gene p.I27L SNP was modestly associated with having early-onset, MODY-like diabetes in the Turkish population. HNF1A gene p.I27L SNP might contribute to age at diabetes diagnosis and family inheritance.
Collapse
Affiliation(s)
- Selvihan Beysel
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Teaching and Training Research Hospital, Ankara, Turkey
- Department of Medical Biology, Baskent University, Ankara, Turkey
- Department of Endocrinology and Metabolism, Afyonkarahisar Saglik Bilimleri University, Afyonkarahisar, Turkey
| | - Nilnur Eyerci
- Department of Genetic Research, Diskapi Yildirim Beyazit Teaching and Research Hospital, Ankara, Turkey
| | - Ferda Alparslan Pinarli
- Department of Genetic Research, Diskapi Yildirim Beyazit Teaching and Research Hospital, Ankara, Turkey
| | - Muhammed Kizilgul
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Teaching and Training Research Hospital, Ankara, Turkey
| | - Ozgur Ozcelik
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Teaching and Training Research Hospital, Ankara, Turkey
| | - Mustafa Caliskan
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Teaching and Training Research Hospital, Ankara, Turkey
| | - Erman Cakal
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Teaching and Training Research Hospital, Ankara, Turkey
| |
Collapse
|
152
|
Pancreatic islet chromatin accessibility and conformation reveals distal enhancer networks of type 2 diabetes risk. Nat Commun 2019; 10:2078. [PMID: 31064983 PMCID: PMC6505525 DOI: 10.1038/s41467-019-09975-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Genetic variants affecting pancreatic islet enhancers are central to T2D risk, but the gene targets of islet enhancer activity are largely unknown. We generate a high-resolution map of islet chromatin loops using Hi-C assays in three islet samples and use loops to annotate target genes of islet enhancers defined using ATAC-seq and published ChIP-seq data. We identify candidate target genes for thousands of islet enhancers, and find that enhancer looping is correlated with islet-specific gene expression. We fine-map T2D risk variants affecting islet enhancers, and find that candidate target genes of these variants defined using chromatin looping and eQTL mapping are enriched in protein transport and secretion pathways. At IGF2BP2, a fine-mapped T2D variant reduces islet enhancer activity and IGF2BP2 expression, and conditional inactivation of IGF2BP2 in mouse islets impairs glucose-stimulated insulin secretion. Our findings provide a resource for studying islet enhancer function and identifying genes involved in T2D risk. Risk loci for type 2 diabetes (T2D) reside in pancreatic islet enhancers. Here, the authors generate high-resolution maps of islet chromatin conformation using Hi-C which they combine with ATAC-seq and ChIP-seq data to annotate candidate target genes of enhancers and validate IGF2BP2 activity in mouse islets.
Collapse
|
153
|
Peng H, Zhu Y, Goldberg J, Vaccarino V, Zhao J. DNA Methylation of Five Core Circadian Genes Jointly Contributes to Glucose Metabolism: A Gene-Set Analysis in Monozygotic Twins. Front Genet 2019; 10:329. [PMID: 31031806 PMCID: PMC6473046 DOI: 10.3389/fgene.2019.00329] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/28/2019] [Indexed: 02/06/2023] Open
Abstract
The timing of daily fluctuations in blood glucose is tightly controlled by the circadian rhythm. DNA methylation accompanies the circadian clock, and aberrant DNA methylation has been associated with circadian disruption and hyperglycemia. However, the precise role of circadian genes methylation in glucose metabolism is unknown. Using a gene-set approach in monozygotic (MZ) twin pairs, we examined the joint effect of 77 CpGs in five core circadian genes (CLOCK, BMAL1, PER1, PER2, PER3) on glucose-related traits in 138 middle-aged, male-male MZ twins (69 pairs). DNA methylation was quantified by bisulfite pyrosequencing. We first conducted matched twin pair analysis to examine the association of single CpG methylation with glucose metabolism. We then performed gene-based and gene-set analyses by the truncated product method to examine the combined effect of DNA methylation at multiple CpGs in a gene or all five circadian genes as a pathway on glucose metabolism. Of the 77 assayed CpGs, only one site was individually associated with insulin resistance at FDR < 0.05. However, the joint effect of DNA methylation in all five circadian genes together showed a significant association with glucose metabolism. Our results may unravel a biological mechanism through which circadian rhythm regulates blood glucose, and highlight the importance of testing the joint effect of multiple CpGs in epigenetic analysis.
Collapse
Affiliation(s)
- Hao Peng
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, United States
| | - Yun Zhu
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, United States
| | - Jack Goldberg
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
154
|
Bentley AR, Sung YJ, Brown MR, Winkler TW, Kraja AT, Ntalla I, Schwander K, Chasman DI, Lim E, Deng X, Guo X, Liu J, Lu Y, Cheng CY, Sim X, Vojinovic D, Huffman JE, Musani SK, Li C, Feitosa MF, Richard MA, Noordam R, Baker J, Chen G, Aschard H, Bartz TM, Ding J, Dorajoo R, Manning AK, Rankinen T, Smith AV, Tajuddin SM, Zhao W, Graff M, Alver M, Boissel M, Chai JF, Chen X, Divers J, Evangelou E, Gao C, Goel A, Hagemeijer Y, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Hung YJ, Jackson AU, Kasturiratne A, Komulainen P, Kühnel B, Leander K, Lin KH, Luan J, Lyytikäinen LP, Matoba N, Nolte IM, Pietzner M, Prins B, Riaz M, Robino A, Said MA, Schupf N, Scott RA, Sofer T, Stancáková A, Takeuchi F, Tayo BO, van der Most PJ, Varga TV, Wang TD, Wang Y, Ware EB, Wen W, Xiang YB, Yanek LR, Zhang W, Zhao JH, Adeyemo A, Afaq S, Amin N, Amini M, Arking DE, Arzumanyan Z, Aung T, Ballantyne C, Barr RG, Bielak LF, Boerwinkle E, Bottinger EP, Broeckel U, Brown M, Cade BE, Campbell A, Canouil M, Charumathi S, Chen YDI, Christensen K, et alBentley AR, Sung YJ, Brown MR, Winkler TW, Kraja AT, Ntalla I, Schwander K, Chasman DI, Lim E, Deng X, Guo X, Liu J, Lu Y, Cheng CY, Sim X, Vojinovic D, Huffman JE, Musani SK, Li C, Feitosa MF, Richard MA, Noordam R, Baker J, Chen G, Aschard H, Bartz TM, Ding J, Dorajoo R, Manning AK, Rankinen T, Smith AV, Tajuddin SM, Zhao W, Graff M, Alver M, Boissel M, Chai JF, Chen X, Divers J, Evangelou E, Gao C, Goel A, Hagemeijer Y, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Hung YJ, Jackson AU, Kasturiratne A, Komulainen P, Kühnel B, Leander K, Lin KH, Luan J, Lyytikäinen LP, Matoba N, Nolte IM, Pietzner M, Prins B, Riaz M, Robino A, Said MA, Schupf N, Scott RA, Sofer T, Stancáková A, Takeuchi F, Tayo BO, van der Most PJ, Varga TV, Wang TD, Wang Y, Ware EB, Wen W, Xiang YB, Yanek LR, Zhang W, Zhao JH, Adeyemo A, Afaq S, Amin N, Amini M, Arking DE, Arzumanyan Z, Aung T, Ballantyne C, Barr RG, Bielak LF, Boerwinkle E, Bottinger EP, Broeckel U, Brown M, Cade BE, Campbell A, Canouil M, Charumathi S, Chen YDI, Christensen K, Concas MP, Connell JM, de las Fuentes L, de Silva HJ, de Vries PS, Doumatey A, Duan Q, Eaton CB, Eppinga RN, Faul JD, Floyd JS, Forouhi NG, Forrester T, Friedlander Y, Gandin I, Gao H, Ghanbari M, Gharib SA, Gigante B, Giulianini F, Grabe HJ, Gu CC, Harris TB, Heikkinen S, Heng CK, Hirata M, Hixson JE, Ikram MA, Jia Y, Joehanes R, Johnson C, Jonas JB, Justice AE, Katsuya T, Khor CC, Kilpeläinen TO, Koh WP, Kolcic I, Kooperberg C, Krieger JE, Kritchevsky SB, Kubo M, Kuusisto J, Lakka TA, Langefeld CD, Langenberg C, Launer LJ, Lehne B, Lewis CE, Li Y, Liang J, Lin S, Liu CT, Liu J, Liu K, Loh M, Lohman KK, Louie T, Luzzi A, Mägi R, Mahajan A, Manichaikul AW, McKenzie CA, Meitinger T, Metspalu A, Milaneschi Y, Milani L, Mohlke KL, Momozawa Y, Morris AP, Murray AD, Nalls MA, Nauck M, Nelson CP, North KE, O'Connell JR, Palmer ND, Papanicolau GJ, Pedersen NL, Peters A, Peyser PA, Polasek O, Poulter N, Raitakari OT, Reiner AP, Renström F, Rice TK, Rich SS, Robinson JG, Rose LM, Rosendaal FR, Rudan I, Schmidt CO, Schreiner PJ, Scott WR, Sever P, Shi Y, Sidney S, Sims M, Smith JA, Snieder H, Starr JM, Strauch K, Stringham HM, Tan NYQ, Tang H, Taylor KD, Teo YY, Tham YC, Tiemeier H, Turner ST, Uitterlinden AG, van Heemst D, Waldenberger M, Wang H, Wang L, Wang L, Wei WB, Williams CA, Wilson G, Wojczynski MK, Yao J, Young K, Yu C, Yuan JM, Zhou J, Zonderman AB, Becker DM, Boehnke M, Bowden DW, Chambers JC, Cooper RS, de Faire U, Deary IJ, Elliott P, Esko T, Farrall M, Franks PW, Freedman BI, Froguel P, Gasparini P, Gieger C, Horta BL, Juang JMJ, Kamatani Y, Kammerer CM, Kato N, Kooner JS, Laakso M, Laurie CC, Lee IT, Lehtimäki T, Magnusson PKE, Oldehinkel AJ, Penninx BWJH, Pereira AC, Rauramaa R, Redline S, Samani NJ, Scott J, Shu XO, van der Harst P, Wagenknecht LE, Wang JS, Wang YX, Wareham NJ, Watkins H, Weir DR, Wickremasinghe AR, Wu T, Zeggini E, Zheng W, Bouchard C, Evans MK, Gudnason V, Kardia SLR, Liu Y, Psaty BM, Ridker PM, van Dam RM, Mook-Kanamori DO, Fornage M, Province MA, Kelly TN, Fox ER, Hayward C, van Duijn CM, Tai ES, Wong TY, Loos RJF, Franceschini N, Rotter JI, Zhu X, Bierut LJ, Gauderman WJ, Rice K, Munroe PB, Morrison AC, Rao DC, Rotimi CN, Cupples LA. Multi-ancestry genome-wide gene-smoking interaction study of 387,272 individuals identifies new loci associated with serum lipids. Nat Genet 2019; 51:636-648. [PMID: 30926973 PMCID: PMC6467258 DOI: 10.1038/s41588-019-0378-y] [Show More Authors] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/07/2019] [Indexed: 12/08/2022]
Abstract
The concentrations of high- and low-density-lipoprotein cholesterol and triglycerides are influenced by smoking, but it is unknown whether genetic associations with lipids may be modified by smoking. We conducted a multi-ancestry genome-wide gene-smoking interaction study in 133,805 individuals with follow-up in an additional 253,467 individuals. Combined meta-analyses identified 13 new loci associated with lipids, some of which were detected only because association differed by smoking status. Additionally, we demonstrate the importance of including diverse populations, particularly in studies of interactions with lifestyle factors, where genomic and lifestyle differences by ancestry may contribute to novel findings.
Collapse
Affiliation(s)
- Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA.
| | - Yun J Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ioanna Ntalla
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jingmin Liu
- Women's Health Initiative Clinical Coordinating Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yingchang Lu
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Quantitative Medicine, Academic Medicine Research Institute, Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jennifer E Huffman
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, GA, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Melissa A Richard
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Raymond Noordam
- Internal Medicine, Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jenna Baker
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Hugues Aschard
- Centre de Bioinformatique, Biostatistique, et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Biostatistics, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jingzhong Ding
- Center on Diabetes, Obesity, and Metabolism, Gerontology and Geriatric Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mathilde Boissel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jasmin Divers
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Yanick Hagemeijer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Fernando P Hartwig
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andrea R V R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yi-Jen Hung
- Endocrinology and Metabolism, Tri-Service General Hospital, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Pirjo Komulainen
- Foundation for Research in Health, Exercise, and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Keng-Hung Lin
- Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jian'an Luan
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Technology, Tampere University, Tampere, Finland
| | - Nana Matoba
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ilja M Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Maik Pietzner
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Bram Prins
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Muhammad Riaz
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Antonietta Robino
- Institute for Maternal and Child Health, IRCCS 'Burlo Garofolo', Trieste, Italy
| | - M Abdullah Said
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Robert A Scott
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Tamar Sofer
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Alena Stancáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Peter J van der Most
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Tzung-Dau Wang
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
- National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yajuan Wang
- Department of Population Quantitative and Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yong-Bing Xiang
- SKLORG and Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Weihua Zhang
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
| | - Jing Hua Zhao
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Saima Afaq
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marzyeh Amini
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zorayr Arzumanyan
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Christie Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Houston Methodist Debakey Heart and Vascular Center, Houston, TX, USA
| | - R Graham Barr
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Erwin P Bottinger
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Departments of Pediatrics, Medicine, and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Morris Brown
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Mickaël Canouil
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
| | - Sabanayagam Charumathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Quantitative Medicine, Academic Medicine Research Institute, Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kaare Christensen
- Danish Aging Research Center, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Maria Pina Concas
- Institute for Maternal and Child Health, IRCCS 'Burlo Garofolo', Trieste, Italy
| | - John M Connell
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ayo Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles B Eaton
- Department of Family Medicine and Epidemiology, Alpert Medical School of Brown University, Providence, RI, USA
| | - Ruben N Eppinga
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Terrence Forrester
- UWI Solutions for Developing Countries, University of the West Indies, Kingston, Jamaica
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - He Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, US National Institutes of Health, Bethesda, MD, USA
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Minato-ku, Japan
| | - James E Hixson
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yucheng Jia
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Roby Joehanes
- Hebrew SeniorLife, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Jost Bruno Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
| | - Anne E Justice
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Ivana Kolcic
- Department of Public Health, Department of Medicine, University of Split, Split, Croatia
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Timo A Lakka
- Foundation for Research in Health, Exercise, and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Claudia Langenberg
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, US National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Lehne
- Institute of Clinical Sciences, Department of Molecular Sciences, Imperial College, London, UK
| | - Cora E Lewis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yize Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingjing Liang
- Department of Population Quantitative and Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Shiow Lin
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kiang Liu
- Epidemiology, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marie Loh
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kurt K Lohman
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Anna Luzzi
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Colin A McKenzie
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research, University of the West Indies, Mona, Jamaica
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Technische Universität München, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Andrew P Morris
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Alison D Murray
- Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - George J Papanicolau
- Epidemiology Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Epidemiology, Faculty of Medicine, Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig Maximilian University, Munich, Germany
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ozren Polasek
- Department of Public Health, Department of Medicine, University of Split, Split, Croatia
- Psychiatric Hospital 'Sveti Ivan', Zagreb, Croatia
- Gen-info, Ltd, Zagreb, Croatia
| | - Neil Poulter
- School of Public Health, Imperial College, London, UK
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Department of Biobank Research, Umeå University, Umeå, Sweden
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jennifer G Robinson
- Department of Epidemiology and Medicine, University of Iowa, Iowa City, IA, USA
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Frits R Rosendaal
- Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Carsten O Schmidt
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - William R Scott
- Institute of Clinical Sciences, Department of Molecular Sciences, Imperial College, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Peter Sever
- National Heart and Lung Institute, Imperial College, London, UK
| | - Yuan Shi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Stephen Sidney
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Konstantin Strauch
- Genetic Epidemiology, Faculty of Medicine, Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig Maximilian University, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Y Q Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Hua Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Social and Behavioral Sciences, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Diana van Heemst
- Internal Medicine, Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Heming Wang
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Lan Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Wen Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Christine A Williams
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory Wilson
- Jackson Heart Study, School of Public Health, Jackson State University, Jackson, MS, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kristin Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Bowden
- Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imperial College Healthcare NHS Trust, London, UK
| | - Richard S Cooper
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Psychology, University of Edinburgh, Edinburgh, UK
| | - Paul Elliott
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- OCDEM, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Barry I Freedman
- Nephrology, Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
- Department of Genomics of Common Disease, Imperial College, London, UK
| | - Paolo Gasparini
- Institute for Maternal and Child Health, IRCCS 'Burlo Garofolo', Trieste, Italy
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Bernardo L Horta
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Jyh-Ming Jimmy Juang
- National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Candace M Kammerer
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jaspal S Kooner
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- National Heart and Lung Institute, Imperial College, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - I-Te Lee
- Endocrinology and Metabolism, Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Technology, Tampere University, Tampere, Finland
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Albertine J Oldehinkel
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Rainer Rauramaa
- Foundation for Research in Health, Exercise, and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - James Scott
- National Heart and Lung Institute, Imperial College, London, UK
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jun-Sing Wang
- Endocrinology and Metabolism, Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | | | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Eleftheria Zeggini
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dennis O Mook-Kanamori
- Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Tanika N Kelly
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Ervin R Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiaofeng Zhu
- Department of Population Quantitative and Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Laura J Bierut
- Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - W James Gauderman
- Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA.
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
- Framingham Heart Study, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
155
|
Marott SCW, Nordestgaard BG, Tybjærg-Hansen A, Benn M. Causal Associations in Type 2 Diabetes Development. J Clin Endocrinol Metab 2019; 104:1313-1324. [PMID: 30566658 DOI: 10.1210/jc.2018-01648] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022]
Abstract
CONTEXT Obesity, glucose, insulin resistance [homeostatic model assessment, version 2, for insulin resistance (HOMA2-IR)], and insulin secretion (HOMA2-β) have been associated with type 2 diabetes (T2D) observationally. However, the causal, genetic contribution of each parameter to this risk is largely unknown and important to study because observational data are prone to confounding but genetic, causal data are free of confounding and reverse causation. OBJECTIVE We examined the causal, genetic contribution of body mass index (BMI), glucose level, C-peptide level, HOMA2-IR, and HOMA2-β to the risk of T2D in 95,540 individuals from the Copenhagen General Population Study and estimated the absolute 10-year risks. METHODS Cox regression analysis, instrumental variable analysis, and Poisson regression analysis were performed to estimate the observational hazard ratios, causal, genetic ORs, and absolute 10-year risks of T2D. RESULTS For 1-SD greater level, BMI was associated with an observational 66% (95% CI, 62% to 72%) and causal, genetic 121% (95% CI, 25% to 291%) greater risk of T2D; glucose with an observational 44% (95% CI, 41% to 46%) and causal, genetic 183% (95% CI, 56% to 416%) greater risk of T2D; and HOMA2-IR with an observational 30% (95% CI, 18% to 44%) and causal, genetic 12% (95% CI, 2% to 22%) greater risk of T2D. In contrast, for 1-SD greater level, HOMA2-β was associated with an observational 14% (95% CI, 11% to 16%) and causal, genetic 21% (95% CI, 8% to 32%) lower risk of T2D. The upper tertiles of HOMA2-IR were associated with absolute 10-year diabetes risks of 31% and 37% in obese women and men, age >60 years, and a glucose level of 6.1 to 11.0 mmol/L. CONCLUSIONS BMI, glucose level, HOMA2-IR, and HOMA2-β are causally associated with T2D.
Collapse
Affiliation(s)
- Sarah C W Marott
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Marianne Benn
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| |
Collapse
|
156
|
Fernández-Tajes J, Gaulton KJ, van de Bunt M, Torres J, Thurner M, Mahajan A, Gloyn AL, Lage K, McCarthy MI. Developing a network view of type 2 diabetes risk pathways through integration of genetic, genomic and functional data. Genome Med 2019; 11:19. [PMID: 30914061 PMCID: PMC6436236 DOI: 10.1186/s13073-019-0628-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 03/08/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified several hundred susceptibility loci for type 2 diabetes (T2D). One critical, but unresolved, issue concerns the extent to which the mechanisms through which these diverse signals influencing T2D predisposition converge on a limited set of biological processes. However, the causal variants identified by GWAS mostly fall into a non-coding sequence, complicating the task of defining the effector transcripts through which they operate. METHODS Here, we describe implementation of an analytical pipeline to address this question. First, we integrate multiple sources of genetic, genomic and biological data to assign positional candidacy scores to the genes that map to T2D GWAS signals. Second, we introduce genes with high scores as seeds within a network optimization algorithm (the asymmetric prize-collecting Steiner tree approach) which uses external, experimentally confirmed protein-protein interaction (PPI) data to generate high-confidence sub-networks. Third, we use GWAS data to test the T2D association enrichment of the "non-seed" proteins introduced into the network, as a measure of the overall functional connectivity of the network. RESULTS We find (a) non-seed proteins in the T2D protein-interaction network so generated (comprising 705 nodes) are enriched for association to T2D (p = 0.0014) but not control traits, (b) stronger T2D-enrichment for islets than other tissues when we use RNA expression data to generate tissue-specific PPI networks and (c) enhanced enrichment (p = 3.9 × 10- 5) when we combine the analysis of the islet-specific PPI network with a focus on the subset of T2D GWAS loci which act through defective insulin secretion. CONCLUSIONS These analyses reveal a pattern of non-random functional connectivity between candidate causal genes at T2D GWAS loci and highlight the products of genes including YWHAG, SMAD4 or CDK2 as potential contributors to T2D-relevant islet dysfunction. The approach we describe can be applied to other complex genetic and genomic datasets, facilitating integration of diverse data types into disease-associated networks.
Collapse
Affiliation(s)
- Juan Fernández-Tajes
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kyle J. Gaulton
- 0000 0001 2107 4242grid.266100.3Department of Pediatrics, University of California, San Diego, CA USA
| | - Martijn van de Bunt
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,0000 0004 1936 8948grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK ,Present Address: Department of Bioinformatics and Data Mining, Novo Nordisk A/S, Maaloev, Denmark
| | - Jason Torres
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,0000 0004 1936 8948grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Matthias Thurner
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,0000 0004 1936 8948grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Anubha Mahajan
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Anna L. Gloyn
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,0000 0004 1936 8948grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK ,0000 0004 0488 9484grid.415719.fOxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Kasper Lage
- 0000 0004 0386 9924grid.32224.35Department of Surgery, Massachusetts, General Hospital, Boston, MA USA ,grid.66859.34Broad Institute of MIT and Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - Mark I. McCarthy
- 0000 0004 1936 8948grid.4991.5Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,0000 0004 1936 8948grid.4991.5Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK ,0000 0004 0488 9484grid.415719.fOxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
157
|
Barroso I, McCarthy MI. The Genetic Basis of Metabolic Disease. Cell 2019; 177:146-161. [PMID: 30901536 PMCID: PMC6432945 DOI: 10.1016/j.cell.2019.02.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Recent developments in genetics and genomics are providing a detailed and systematic characterization of the genetic underpinnings of common metabolic diseases and traits, highlighting the inherent complexity within systems for homeostatic control and the many ways in which that control can fail. The genetic architecture underlying these common metabolic phenotypes is complex, with each trait influenced by hundreds of loci spanning a range of allele frequencies and effect sizes. Here, we review the growing appreciation of this complexity and how this has fostered the implementation of genome-scale approaches that deliver robust mechanistic inference and unveil new strategies for translational exploitation.
Collapse
Affiliation(s)
- Inês Barroso
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| |
Collapse
|
158
|
The Homeodomain Transcription Factor NKX2.1 Is Essential for the Early Specification of Melanocortin Neuron Identity and Activates Pomc Expression in the Developing Hypothalamus. J Neurosci 2019; 39:4023-4035. [PMID: 30886014 DOI: 10.1523/jneurosci.2924-18.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022] Open
Abstract
Food intake is tightly regulated by a group of neurons present in the arcuate nucleus of the hypothalamus, which release Pomc-encoded melanocortins, the absence of which induces marked hyperphagia and early-onset obesity. Although the relevance of hypothalamic POMC neurons in the regulation of body weight and energy balance is well appreciated, little is known about the transcription factors that establish the melanocortin neuron identity during brain development and its phenotypic maintenance in postnatal life. Here, we report that the transcription factor NKX2.1 is present in mouse hypothalamic POMC neurons from early development to adulthood. Electromobility shift assays showed that NKX2.1 binds in vitro to NKX binding motifs present in the neuronal Pomc enhancers nPE1 and nPE2 and chromatin immunoprecipitation assays detected in vivo binding of NKX2.1 to nPE1 and nPE2 in mouse hypothalamic extracts. Transgenic and mutant studies performed in mouse embryos of either sex and adult males showed that the NKX motifs present in nPE1 and nPE2 are essential for their transcriptional enhancer activity. The conditional early inactivation of Nkx2.1 in the ventral hypothalamus prevented the onset of Pomc expression. Selective Nkx2.1 ablation from POMC neurons decreased Pomc expression in adult males and mildly increased their body weight and adiposity. Our results demonstrate that NKX2.1 is necessary to activate Pomc expression by binding to conserved canonical NKX motifs present in nPE1 and nPE2. Therefore, NKX2.1 plays a critical role in the early establishment of hypothalamic melanocortin neuron identity and participates in the maintenance of Pomc expression levels during adulthood.SIGNIFICANCE STATEMENT Food intake and body weight regulation depend on hypothalamic neurons that release satiety-inducing neuropeptides, known as melanocortins. Central melanocortins are encoded byPomc, and Pomc mutations may lead to hyperphagia and severe obesity. Although the importance of central melanocortins is well appreciated, the genetic program that establishes and maintains fully functional POMC neurons remains to be explored. Here, we combined molecular, genetic, developmental, and functional studies that led to the discovery of NKX2.1, a transcription factor that participates in the early morphogenesis of the developing hypothalamus, as a key player in establishing the early identity of melanocortin neurons by activating Pomc expression. Thus, Nkx2.1 adds to the growing list of genes that participate in body weight regulation and adiposity.
Collapse
|
159
|
Hébert HL, Shepherd B, Milburn K, Veluchamy A, Meng W, Carr F, Donnelly LA, Tavendale R, Leese G, Colhoun HM, Dow E, Morris AD, Doney AS, Lang CC, Pearson ER, Smith BH, Palmer CNA. Cohort Profile: Genetics of Diabetes Audit and Research in Tayside Scotland (GoDARTS). Int J Epidemiol 2019; 47:380-381j. [PMID: 29025058 PMCID: PMC5913637 DOI: 10.1093/ije/dyx140] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
| | | | - Keith Milburn
- Health Informatics Centre Services, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Abirami Veluchamy
- Division of Population Health Sciences.,Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | | | - Fiona Carr
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | | | - Roger Tavendale
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | - Graham Leese
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | - Helen M Colhoun
- Division of Population Health Sciences.,Institute of Genetics & Molecular Medicine
| | - Ellie Dow
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | - Andrew D Morris
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | | | - Chim C Lang
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | - Ewan R Pearson
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics
| | | | | |
Collapse
|
160
|
Sieber KB, Batorsky A, Siebenthall K, Hudkins KL, Vierstra JD, Sullivan S, Sur A, McNulty M, Sandstrom R, Reynolds A, Bates D, Diegel M, Dunn D, Nelson J, Buckley M, Kaul R, Sampson MG, Himmelfarb J, Alpers CE, Waterworth D, Akilesh S. Integrated Functional Genomic Analysis Enables Annotation of Kidney Genome-Wide Association Study Loci. J Am Soc Nephrol 2019; 30:421-441. [PMID: 30760496 PMCID: PMC6405142 DOI: 10.1681/asn.2018030309] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 12/26/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Linking genetic risk loci identified by genome-wide association studies (GWAS) to their causal genes remains a major challenge. Disease-associated genetic variants are concentrated in regions containing regulatory DNA elements, such as promoters and enhancers. Although researchers have previously published DNA maps of these regulatory regions for kidney tubule cells and glomerular endothelial cells, maps for podocytes and mesangial cells have not been available. METHODS We generated regulatory DNA maps (DNase-seq) and paired gene expression profiles (RNA-seq) from primary outgrowth cultures of human glomeruli that were composed mainly of podocytes and mesangial cells. We generated similar datasets from renal cortex cultures, to compare with those of the glomerular cultures. Because regulatory DNA elements can act on target genes across large genomic distances, we also generated a chromatin conformation map from freshly isolated human glomeruli. RESULTS We identified thousands of unique regulatory DNA elements, many located close to transcription factor genes, which the glomerular and cortex samples expressed at different levels. We found that genetic variants associated with kidney diseases (GWAS) and kidney expression quantitative trait loci were enriched in regulatory DNA regions. By combining GWAS, epigenomic, and chromatin conformation data, we functionally annotated 46 kidney disease genes. CONCLUSIONS We demonstrate a powerful approach to functionally connect kidney disease-/trait-associated loci to their target genes by leveraging unique regulatory DNA maps and integrated epigenomic and genetic analysis. This process can be applied to other kidney cell types and will enhance our understanding of genome regulation and its effects on gene expression in kidney disease.
Collapse
Affiliation(s)
| | - Anna Batorsky
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | | | | | - Jeff D Vierstra
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | | | - Aakash Sur
- Phase Genomics Inc., Seattle, Washington
- Department of Biomedical and Health Informatics, and
| | - Michelle McNulty
- Division of Pediatric Nephrology, Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan; and
| | | | - Alex Reynolds
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Daniel Bates
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Morgan Diegel
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Douglass Dunn
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Jemma Nelson
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Michael Buckley
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Rajinder Kaul
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan; and
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Charles E Alpers
- Department of Anatomic Pathology
- Kidney Research Institute, Seattle, Washington
| | | | - Shreeram Akilesh
- Department of Anatomic Pathology,
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
161
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to summarize recent advances in the genomics of type 2 diabetes (T2D) and to highlight current initiatives to advance precision health. RECENT FINDINGS Generation of multi-omic data to measure each of the "biologic layers," developments in describing genomic function and annotation in T2D relevant tissue, along with the increasing recognition that T2D is a heterogeneous disease, and large-scale collaborations have all contributed to advancing our understanding of the molecular basis of T2D. Substantial advances have been made in understanding the molecular basis of T2D pathogenesis, such that precision health diabetes is increasingly becoming a reality. For precision diabetes to become a routine in clinical and public health, additional large-scale multi-omic initiatives are needed along with better assessment of our environment to delineate an individual's diabetes subtype for improved detection and management.
Collapse
Affiliation(s)
- Yuan Lin
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| | - Jennifer Wessel
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA.
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Diabetes Translational Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
162
|
Haljas K, Hakaste L, Lahti J, Isomaa B, Groop L, Tuomi T, Räikkönen K. The associations of daylight and melatonin receptor 1B gene rs10830963 variant with glycemic traits: the prospective PPP-Botnia study. Ann Med 2019; 51:58-67. [PMID: 30592226 PMCID: PMC7857441 DOI: 10.1080/07853890.2018.1564357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Seasonal variation in glucose metabolism might be driven by changes in daylight. Melatonin entrains circadian regulation and is directly associated with daylight. The relationship between melatonin receptor 1B gene variants with glycemic traits and type 2 diabetes is well established. We studied if daylight length was associated with glycemic traits and if it modified the relationship between melatonin receptor 1B gene rs10830963 variant and glycemic traits. MATERIALS A population-based sample of 3422 18-78-year-old individuals without diabetes underwent an oral glucose tolerance test twice, an average 6.8 years (SD = 0.9) apart and were genotyped for rs10830963. Daylight data was obtained from the Finnish Meteorological Institute. RESULTS Cross-sectionally, more daylight was associated with lower fasting glucose, but worse insulin sensitivity and secretion at follow-up. Longitudinally, individuals studied on lighter days at follow-up than at baseline showed higher glucose values during the oral glucose tolerance test and lower Corrected Insulin Response at follow-up. GG genotype carriers in the rs10830963 became more insulin resistant during follow-up if daylight length was shorter at follow-up than at baseline. CONCLUSIONS Our study shows that individual glycemic profiles may vary according to daylight, MTNR1B genotype and their interaction. Future studies may consider taking daylight length into account. Key messages In Western Finland, the amount daylight follows an extensive annual variation ranging from 4 h 44 min to 20 h 17 min, making it ideal to study the associations between daylight and glycemic traits. Moreover, this allows researchers to explore if the relationship between the melatonin receptor 1B gene rs10830963 variant and glycemic traits is modified by the amount of daylight both cross-sectionally and longitudinally. This study shows that individuals, who participated in the study on lighter days at the follow-up than at the baseline, displayed to a greater extent worse glycemic profiles across the follow-up. Novel findings from the current study show that in the longitudinal analyses, each addition of the minor G allele of the melatonin receptor 1B gene rs10830963 was associated with worsening of fasting glucose values and insulin secretion across the 6.8-year follow-up. Importantly, this study shows that in those with the rs10830963 GG genotype, insulin sensitivity deteriorated the most significantly across the 6.8-year follow-up if the daylight length on the oral glucose tolerance testing date at the follow-up was shorter than at the baseline. Taken together, the current findings suggest that the amount of daylight may affect glycemic traits, especially fasting glucose and insulin secretion even though the effect size is small. The association can very according to the rs10830963 risk variant. Further research is needed to elucidate the mechanisms behind these associations.
Collapse
Affiliation(s)
- Kadri Haljas
- a Department of Psychology and Logopedics , University of Helsinki , Helsinki , Finland
| | - Liisa Hakaste
- b Department of Endocrinology, Abdominal Centre , Helsinki University Hospital , Helsinki , Finland.,c Folkhälsan Research Center , Helsinki , Finland
| | - Jari Lahti
- a Department of Psychology and Logopedics , University of Helsinki , Helsinki , Finland.,d Helsinki Collegium for Advanced Studies , University of Helsinki , Helsinki , Finland
| | - Bo Isomaa
- c Folkhälsan Research Center , Helsinki , Finland.,e Department of Social Services and Health Care , Jakobstad , Finland
| | - Leif Groop
- f Finnish Institute for Molecular Medicine, University of Helsinki , Helsinki , Finland.,g Department of Clinical Sciences, Diabetes and Endocrinology , Lund University , Malmö , Sweden
| | - Tiinamaija Tuomi
- b Department of Endocrinology, Abdominal Centre , Helsinki University Hospital , Helsinki , Finland.,c Folkhälsan Research Center , Helsinki , Finland.,f Finnish Institute for Molecular Medicine, University of Helsinki , Helsinki , Finland
| | - Katri Räikkönen
- a Department of Psychology and Logopedics , University of Helsinki , Helsinki , Finland
| |
Collapse
|
163
|
Abstract
Despite considerable advances in the past few years, obesity and type 2 diabetes mellitus (T2DM) remain two major challenges for public health systems globally. In the past 9 years, genome-wide association studies (GWAS) have established a major role for genetic variation within the MTNR1B locus in regulating fasting plasma levels of glucose and in affecting the risk of T2DM. This discovery generated a major interest in the melatonergic system, in particular the melatonin MT2 receptor (which is encoded by MTNR1B). In this Review, we discuss the effect of melatonin and its receptors on glucose homeostasis, obesity and T2DM. Preclinical and clinical post-GWAS evidence of frequent and rare variants of the MTNR1B locus confirmed its importance in regulating glucose homeostasis and T2DM risk with minor effects on obesity. However, these studies did not solve the question of whether melatonin is beneficial or detrimental, an issue that will be discussed in the context of the peculiarities of the melatonergic system. Melatonin receptors might have therapeutic potential as they belong to the highly druggable G protein-coupled receptor superfamily. Clarifying the precise role of melatonin and its receptors on glucose homeostasis is urgent, as melatonin is widely used for other indications, either as a prescribed medication or as a supplement without medical prescription, in many countries in Europe and in the USA.
Collapse
Affiliation(s)
- Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France.
- CNRS UMR 8104, Paris, France.
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
164
|
Zenin A, Tsepilov Y, Sharapov S, Getmantsev E, Menshikov LI, Fedichev PO, Aulchenko Y. Identification of 12 genetic loci associated with human healthspan. Commun Biol 2019; 2:41. [PMID: 30729179 PMCID: PMC6353874 DOI: 10.1038/s42003-019-0290-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
Aging populations face diminishing quality of life due to increased disease and morbidity. These challenges call for longevity research to focus on understanding the pathways controlling healthspan. We use the data from the UK Biobank (UKB) cohort and observe that the risks of major chronic diseases increased exponentially and double every eight years, i.e., at a rate compatible with the Gompertz mortality law. Assuming that aging drives the acceleration in morbidity rates, we build a risk model to predict the age at the end of healthspan depending on age, gender, and genetic background. Using the sub-population of 300,447 British individuals as a discovery cohort, we identify 12 loci associated with healthspan at the whole-genome significance level. We find strong genetic correlations between healthspan and all-cause mortality, life-history, and lifestyle traits. We thereby conclude that the healthspan offers a promising new way to interrogate the genetics of human longevity.
Collapse
Affiliation(s)
- Aleksandr Zenin
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
| | - Yakov Tsepilov
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
| | - Sodbo Sharapov
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
| | | | - L. I. Menshikov
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
- National Research Center “Kurchatov Institute”, 1, Akademika Kurchatova pl., Moscow, Russia 123182
| | - Peter O. Fedichev
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
- Moscow Institute of Physics and Technology, Institutskii per. 9, Dolgoprudny, Moscow Russia 141700
| | - Yurii Aulchenko
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
- PolyOmica, Het Vlaggeschip 61, 5237PA ‘s-Hertogenbosch, The Netherlands
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, Scotland EH8 9AG UK
| |
Collapse
|
165
|
Zhao J, Feng Q, Wu P, Lupu RA, Wilke RA, Wells QS, Denny JC, Wei WQ. Learning from Longitudinal Data in Electronic Health Record and Genetic Data to Improve Cardiovascular Event Prediction. Sci Rep 2019; 9:717. [PMID: 30679510 PMCID: PMC6345960 DOI: 10.1038/s41598-018-36745-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/23/2018] [Indexed: 02/07/2023] Open
Abstract
Current approaches to predicting a cardiovascular disease (CVD) event rely on conventional risk factors and cross-sectional data. In this study, we applied machine learning and deep learning models to 10-year CVD event prediction by using longitudinal electronic health record (EHR) and genetic data. Our study cohort included 109, 490 individuals. In the first experiment, we extracted aggregated and longitudinal features from EHR. We applied logistic regression, random forests, gradient boosting trees, convolutional neural networks (CNN) and recurrent neural networks with long short-term memory (LSTM) units. In the second experiment, we applied a late-fusion approach to incorporate genetic features. We compared the performance with approaches currently utilized in routine clinical practice - American College of Cardiology and the American Heart Association (ACC/AHA) Pooled Cohort Risk Equation. Our results indicated that incorporating longitudinal feature lead to better event prediction. Combining genetic features through a late-fusion approach can further improve CVD prediction, underscoring the importance of integrating relevant genetic data whenever available.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - QiPing Feng
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Patrick Wu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Roxana A Lupu
- Department of Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Russell A Wilke
- Department of Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Quinn S Wells
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
166
|
Li J, Niu X, Li J, Wang Q. Association of PPARG Gene Polymorphisms Pro12Ala with Type 2 Diabetes Mellitus: A Meta-analysis. Curr Diabetes Rev 2019; 15:277-283. [PMID: 30207237 DOI: 10.2174/1573399814666180912130401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 08/15/2018] [Accepted: 09/04/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Previous studies suggested that the single nucleotide polymorphisms of Pro12Ala located within the PPARG gene were significantly associated with the T2DM. Recently, the genetic studies on Pro12Ala were conducted in the different ethnic groups and the results of each study were shown to be inconsistent. Moreover, the systematic review has not been updated since 2000. OBJECTIVE To further validate the risk of Pro12Ala for T2DM disease based on the genetic data. METHODS The genetic studies on the Pro12Ala in the T2DM were searched in the PubMed and PMC database from January 2000 to October 2017. The meta-analysis was conducted with the CMA software. RESULTS The meta-analysis collected 14 studies including 20702 cases and 36227 controls. The combined analysis of all studies found that Pro12Ala was shown to be significantly associated with T2DM and the Ala allele played the increasing risks for the disease. Nevertheless, publication bias was detected in the combined analysis. The subgroup analysis indicated that Pro12Ala was found to be significant in the Caucasian and Chinese population. There was no heterogeneity and publication bias in these two groups. CONCLUSION The meta-analysis confirmed the evidence that the Pro12Ala was the susceptible variant for the decreasing risks for the T2DM.
Collapse
Affiliation(s)
- Junyan Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Changzhi Medical College Affiliated Heji Hospital, Endocrinology and Metabolism. No. 271, East Taihang Road Changzhi, China
| | - Xiaohong Niu
- Changzhi Medical College Affiliated Heji Hospital, Endocrinology and Metabolism. No. 271, East Taihang Road Changzhi, China
| | - JianBo Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingzhong Wang
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
167
|
Amlie-Wolf A, Tang M, Way J, Dombroski B, Jiang M, Vrettos N, Chou YF, Zhao Y, Kuzma A, Mlynarski EE, Leung YY, Brown CD, Wang LS, Schellenberg GD. Inferring the Molecular Mechanisms of Noncoding Alzheimer's Disease-Associated Genetic Variants. J Alzheimers Dis 2019; 72:301-318. [PMID: 31561366 PMCID: PMC7316086 DOI: 10.3233/jad-190568] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Most of the loci identified by genome-wide association studies (GWAS) for late-onset Alzheimer's disease (LOAD) are in strong linkage disequilibrium (LD) with nearby variants all of which could be the actual functional variants, often in non-protein-coding regions and implicating underlying gene regulatory mechanisms. We set out to characterize the causal variants, regulatory mechanisms, tissue contexts, and target genes underlying these associations. We applied our INFERNO algorithm to the top 19 non-APOE loci from the IGAP GWAS study. INFERNO annotated all LD-expanded variants at each locus with tissue-specific regulatory activity. Bayesian co-localization analysis of summary statistics and eQTL data was performed to identify tissue-specific target genes. INFERNO identified enhancer dysregulation in all 19 tag regions analyzed, significant enrichments of enhancer overlaps in the immune-related blood category, and co-localized eQTL signals overlapping enhancers from the matching tissue class in ten regions (ABCA7, BIN1, CASS4, CD2AP, CD33, CELF1, CLU, EPHA1, FERMT2, ZCWPW1). In several cases, we identified dysregulation of long noncoding RNA (lncRNA) transcripts and applied the lncRNA target identification algorithm from INFERNO to characterize their downstream biological effects. We also validated the allele-specific effects of several variants on enhancer function using luciferase expression assays. By integrating functional genomics with GWAS signals, our analysis yielded insights into the regulatory mechanisms, tissue contexts, genes, and biological processes affected by noncoding genetic variation associated with LOAD risk.
Collapse
Affiliation(s)
- Alexandre Amlie-Wolf
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell Tang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica Way
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beth Dombroski
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Jiang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas Vrettos
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi-Fan Chou
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Zhao
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amanda Kuzma
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elisabeth E. Mlynarski
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuk Yee Leung
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher D. Brown
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li-San Wang
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerard D. Schellenberg
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
168
|
Johansson M, Carreras-Torres R, Scelo G, Purdue MP, Mariosa D, Muller DC, Timpson NJ, Haycock PC, Brown KM, Wang Z, Ye Y, Hofmann JN, Foll M, Gaborieau V, Machiela MJ, Colli LM, Li P, Garnier JG, Blanche H, Boland A, Burdette L, Prokhortchouk E, Skryabin KG, Yeager M, Radojevic-Skodric S, Ognjanovic S, Foretova L, Holcatova I, Janout V, Mates D, Mukeriya A, Rascu S, Zaridze D, Bencko V, Cybulski C, Fabianova E, Jinga V, Lissowska J, Lubinski J, Navratilova M, Rudnai P, Benhamou S, Cancel-Tassin G, Cussenot O, Weiderpass E, Ljungberg B, Tumkur Sitaram R, Häggström C, Bruinsma F, Jordan SJ, Severi G, Winship I, Hveem K, Vatten LJ, Fletcher T, Larsson SC, Wolk A, Banks RE, Selby PJ, Easton DF, Andreotti G, Beane Freeman LE, Koutros S, Männistö S, Weinstein S, Clark PE, Edwards TL, Lipworth L, Gapstur SM, Stevens VL, Carol H, Freedman ML, Pomerantz MM, Cho E, Wilson KM, Gaziano JM, Sesso HD, Freedman ND, Parker AS, Eckel-Passow JE, Huang WY, Kahnoski RJ, Lane BR, Noyes SL, Petillo D, Teh BT, Peters U, White E, Anderson GL, Johnson L, Luo J, Buring J, Lee IM, Chow WH, Moore LE, Eisen T, Henrion M, Larkin J, Barman P, Leibovich BC, et alJohansson M, Carreras-Torres R, Scelo G, Purdue MP, Mariosa D, Muller DC, Timpson NJ, Haycock PC, Brown KM, Wang Z, Ye Y, Hofmann JN, Foll M, Gaborieau V, Machiela MJ, Colli LM, Li P, Garnier JG, Blanche H, Boland A, Burdette L, Prokhortchouk E, Skryabin KG, Yeager M, Radojevic-Skodric S, Ognjanovic S, Foretova L, Holcatova I, Janout V, Mates D, Mukeriya A, Rascu S, Zaridze D, Bencko V, Cybulski C, Fabianova E, Jinga V, Lissowska J, Lubinski J, Navratilova M, Rudnai P, Benhamou S, Cancel-Tassin G, Cussenot O, Weiderpass E, Ljungberg B, Tumkur Sitaram R, Häggström C, Bruinsma F, Jordan SJ, Severi G, Winship I, Hveem K, Vatten LJ, Fletcher T, Larsson SC, Wolk A, Banks RE, Selby PJ, Easton DF, Andreotti G, Beane Freeman LE, Koutros S, Männistö S, Weinstein S, Clark PE, Edwards TL, Lipworth L, Gapstur SM, Stevens VL, Carol H, Freedman ML, Pomerantz MM, Cho E, Wilson KM, Gaziano JM, Sesso HD, Freedman ND, Parker AS, Eckel-Passow JE, Huang WY, Kahnoski RJ, Lane BR, Noyes SL, Petillo D, Teh BT, Peters U, White E, Anderson GL, Johnson L, Luo J, Buring J, Lee IM, Chow WH, Moore LE, Eisen T, Henrion M, Larkin J, Barman P, Leibovich BC, Choueiri TK, Lathrop GM, Deleuze JF, Gunter M, McKay JD, Wu X, Houlston RS, Chanock SJ, Relton C, Richards JB, Martin RM, Davey Smith G, Brennan P. The influence of obesity-related factors in the etiology of renal cell carcinoma-A mendelian randomization study. PLoS Med 2019; 16:e1002724. [PMID: 30605491 PMCID: PMC6317776 DOI: 10.1371/journal.pmed.1002724] [Show More Authors] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several obesity-related factors have been associated with renal cell carcinoma (RCC), but it is unclear which individual factors directly influence risk. We addressed this question using genetic markers as proxies for putative risk factors and evaluated their relation to RCC risk in a mendelian randomization (MR) framework. This methodology limits bias due to confounding and is not affected by reverse causation. METHODS AND FINDINGS Genetic markers associated with obesity measures, blood pressure, lipids, type 2 diabetes, insulin, and glucose were initially identified as instrumental variables, and their association with RCC risk was subsequently evaluated in a genome-wide association study (GWAS) of 10,784 RCC patients and 20,406 control participants in a 2-sample MR framework. The effect on RCC risk was estimated by calculating odds ratios (ORSD) for a standard deviation (SD) increment in each risk factor. The MR analysis indicated that higher body mass index increases the risk of RCC (ORSD: 1.56, 95% confidence interval [CI] 1.44-1.70), with comparable results for waist-to-hip ratio (ORSD: 1.63, 95% CI 1.40-1.90) and body fat percentage (ORSD: 1.66, 95% CI 1.44-1.90). This analysis further indicated that higher fasting insulin (ORSD: 1.82, 95% CI 1.30-2.55) and diastolic blood pressure (DBP; ORSD: 1.28, 95% CI 1.11-1.47), but not systolic blood pressure (ORSD: 0.98, 95% CI 0.84-1.14), increase the risk for RCC. No association with RCC risk was seen for lipids, overall type 2 diabetes, or fasting glucose. CONCLUSIONS This study provides novel evidence for an etiological role of insulin in RCC, as well as confirmatory evidence that obesity and DBP influence RCC risk.
Collapse
Affiliation(s)
| | | | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Mark P. Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Daniela Mariosa
- International Agency for Research on Cancer (IARC), Lyon, France
| | | | - Nicolas J. Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Philip C. Haycock
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Kevin M. Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Zhaoming Wang
- St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Yuanqing Ye
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jonathan N. Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Matthieu Foll
- International Agency for Research on Cancer (IARC), Lyon, France
| | | | - Mitchell J. Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Leandro M. Colli
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Peng Li
- International Agency for Research on Cancer (IARC), Lyon, France
- Max Planck Institute for Demographic Research, Rostock, Germany
| | - Jean-Guillaume Garnier
- Centre National de Genotypage, Institut de Genomique, Centre de l'Energie Atomique et aux Energies Alternatives, Evry, France
- Fondation Jean Dausset - Centre d'Etude du Polymorphisme Humain, Paris, France
| | - Helene Blanche
- Fondation Jean Dausset - Centre d'Etude du Polymorphisme Humain, Paris, France
| | - Anne Boland
- Centre National de Genotypage, Institut de Genomique, Centre de l'Energie Atomique et aux Energies Alternatives, Evry, France
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Egor Prokhortchouk
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Konstantin G. Skryabin
- Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences, Moscow, Russian Federation
- Kurchatov Scientific Center, Moscow, Russian Federation
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Sanja Radojevic-Skodric
- Institute of Pathology, Medical School of Belgrade, Belgrade, Serbia
- Clinic of Urology, Clinical Center of Serbia, Belgrade, Serbia
| | - Simona Ognjanovic
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, United States of America
- International Organization for Cancer Prevention and Research (IOCPR), Belgrade, Serbia
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Ivana Holcatova
- Institute of Public Health and Preventive Medicine, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vladimir Janout
- Department of Preventive Medicine, Faculty of Medicine, Palacky University, Olomouc, Czech Republic
| | - Dana Mates
- National Institute of Public Health, Bucharest, Romania
| | - Anush Mukeriya
- Russian N.N. Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Stefan Rascu
- Carol Davila University of Medicine and Pharmacy, Th. Burghele Hospital, Bucharest, Romania
| | - David Zaridze
- Russian N.N. Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Vladimir Bencko
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Eleonora Fabianova
- Regional Authority of Public Health in Banska Bystrica, Banska Bystrica, Slovakia
| | - Viorel Jinga
- Carol Davila University of Medicine and Pharmacy, Th. Burghele Hospital, Bucharest, Romania
| | - Jolanta Lissowska
- The M Sklodowska-Curie Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Marie Navratilova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Peter Rudnai
- National Public Health Center, National Directorate of Environmental Health, Budapest, Hungary
| | - Simone Benhamou
- INSERM U946, Paris, France
- CNRS UMR8200, Institute Gustave Roussy, Villejuif, France
| | - Geraldine Cancel-Tassin
- CeRePP, Paris, France
- UPMC Univ Paris 06, GRC n°5, Institut Universitaire de Cancérologie, Paris, France
| | - Olivier Cussenot
- CeRePP, Paris, France
- UPMC Univ Paris 06, GRC n°5, Institut Universitaire de Cancérologie, Paris, France
- AP-HP, Department of Urology, Hopitaux Universitaires Est Parisien Tenon, Paris, France
| | - Elisabete Weiderpass
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | | | - Christel Häggström
- Department of Biobank Research, Umeå University, Umeå, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Fiona Bruinsma
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
| | - Susan J. Jordan
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Public Health, The University of Queensland, Brisbane, Australia
| | - Gianluca Severi
- “Health across generations” team, CESP Inserm, Facultés de Médicine Université Paris-Sud, UVSQ, Université Paris-Saclay, Gustave Roussy, Villejuif, France
- Human Genetics Foundation (HuGeF), Torino, Italy
| | - Ingrid Winship
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Kristian Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars J. Vatten
- Department of Public Health and General Practice, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tony Fletcher
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
| | - Susanna C. Larsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rosamonde E. Banks
- Leeds Institute of Cancer and Pathology, University of Leeds, St James's University Hospital, Leeds, United Kingdom
| | - Peter J. Selby
- National Institute for Health Research Diagnostic Evidence Cooperative, Division of Surgery, Imperial College London, St Mary’s Hospital, London, United Kingdom
| | - Douglas F. Easton
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Gabriella Andreotti
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Laura E. Beane Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Satu Männistö
- National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Peter E. Clark
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, United States of America
| | - Todd L. Edwards
- Department of Medicine, Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Vanderbilt Genetics Institute, Nashville, Tennessee, United States of America
| | - Loren Lipworth
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, United States of America
| | - Susan M. Gapstur
- American Cancer Society, Atlanta, Georgia, United States of America
| | | | - Hallie Carol
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Matthew L. Freedman
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Mark M. Pomerantz
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Eunyoung Cho
- Brown University, Providence, Rhode Island, United States of America
| | - Kathryn M. Wilson
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - J. Michael Gaziano
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Howard D. Sesso
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Alexander S. Parker
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Jeanette E. Eckel-Passow
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Richard J. Kahnoski
- Division of Urology, Spectrum Health, Grand Rapids, Michigan, United States of America
| | - Brian R. Lane
- Division of Urology, Spectrum Health, Grand Rapids, Michigan, United States of America
- College of Human Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Sabrina L. Noyes
- Van Andel Research Institute, Center for Cancer Genomics and Quantitative Biology, Grand Rapids, Michigan, United States of America
- Spectrum Health, Grand Rapids, Michigan, United States of America
| | - David Petillo
- Van Andel Research Institute, Center for Cancer Genomics and Quantitative Biology, Grand Rapids, Michigan, United States of America
- Diagnostics Program at Ferris State University, Grand Rapids, Michigan, United States of America
| | - Bin Tean Teh
- Van Andel Research Institute, Center for Cancer Genomics and Quantitative Biology, Grand Rapids, Michigan, United States of America
- Program in Cancer and Stem Cell Biology, Duke-National, University of Singapore Medical School, Singapore, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ulrike Peters
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emily White
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Garnet L. Anderson
- WHI Clinical Coordinating Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lisa Johnson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Juhua Luo
- Department of Epidemiology and Biostatistics, School of Public Health Indiana University Bloomington, Bloomington, Indiana, United States of America
| | - Julie Buring
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - I-Min Lee
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Wong-Ho Chow
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lee E. Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | | | - Marc Henrion
- The Institute of Cancer Research, London, United Kingdom
- Dept. of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - James Larkin
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Poulami Barman
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Bradley C. Leibovich
- Department of Urology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Toni K. Choueiri
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - G. Mark Lathrop
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Jean-Francois Deleuze
- Centre National de Genotypage, Institut de Genomique, Centre de l'Energie Atomique et aux Energies Alternatives, Evry, France
- Fondation Jean Dausset - Centre d'Etude du Polymorphisme Humain, Paris, France
| | - Marc Gunter
- International Agency for Research on Cancer (IARC), Lyon, France
| | - James D. McKay
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Xifeng Wu
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | | | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, Maryland, United States of America
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - J. Brent Richards
- Departments of Medicine, Human Genetics, Epidemiology and Biostatistics, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Richard M. Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- University Hospitals Bristol NHS Foundation Trust National Institute for Health Research Bristol Nutrition Biomedical Research Unit, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
169
|
Zwakenberg SR, Remmelzwaal S, Beulens JWJ, Booth SL, Burgess S, Dashti HS, Imamura F, Feskens EJM, van der Schouw YT, Sluijs I. Circulating Phylloquinone Concentrations and Risk of Type 2 Diabetes: A Mendelian Randomization Study. Diabetes 2019; 68:220-225. [PMID: 30352877 PMCID: PMC6314462 DOI: 10.2337/db18-0543] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 10/12/2018] [Indexed: 01/04/2023]
Abstract
This study investigated the causal relation between circulating phylloquinone (vitamin K1) concentrations and type 2 diabetes by using a Mendelian randomization (MR) approach. We used data from three studies: the European Prospective Investigation into Cancer and Nutrition (EPIC)-InterAct case-cohort study, Diabetes Genetics Replication and Meta-analysis (DIAGRAM), and the UK Biobank, resulting in 69,647 subjects with type 2 diabetes. We calculated a weighted genetic risk score including four genetic variants previously found to be associated with circulating phylloquinone concentrations. Inverse-variance weighted analysis was used to obtain a risk ratio (RR) for the causal relation between circulating phylloquinone concentrations and risk of type 2 diabetes. Presence of pleiotropy and the robustness of the results were assessed using MR-Egger and weighted-median analyses. Genetically predicted concentrations of circulating phylloquinone were associated with lower risk of type 2 diabetes with an RR of 0.93 (95% CI 0.89; 0.97) per every natural logarithm (Ln)-nmol/L-unit increase in circulating phylloquinone. The MR-Egger and weighted median analyses showed RRs of 0.94 (0.86; 1.02) and 0.93 (0.88; 0.98), respectively, indicating no pleiotropy. In conclusion, our study supports that higher circulating phylloquinone may be causally related with lower risk of type 2 diabetes, highlighting the importance of sufficient phylloquinone in the human diet.
Collapse
Affiliation(s)
- Sabine R Zwakenberg
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sharon Remmelzwaal
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Vrije Universiteit University Medical Center, Amsterdam, the Netherlands
| | - Joline W J Beulens
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Vrije Universiteit University Medical Center, Amsterdam, the Netherlands
| | - Sarah L Booth
- Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Stephen Burgess
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, U.K
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, U.K
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Fumiaki Imamura
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, U.K
| | - Edith J M Feskens
- Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
170
|
Li L, Pan Z, Yang X. Identification of dynamic molecular networks in peripheral blood mononuclear cells in type 1 diabetes mellitus. Diabetes Metab Syndr Obes 2019; 12:969-982. [PMID: 31417297 PMCID: PMC6601337 DOI: 10.2147/dmso.s207021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the immune destruction of islet β cells. Gene expression in peripheral blood mononuclear cells (PBMCs) could offer new disease and treatment markers in T1DM. The objective of this study was to explore the coexpression and dynamic molecular networks in PBMCs of T1DM patients. METHODS Dataset GSE9006 contains PBMC samples of healthy volunteers, newly diagnosed T1DM patients, T1DM patients after insulin treatment, and newly diagnosed type 2 diabetes mellitus (T2DM) patients. Weighted correlation network analysis (WGCNA) was used to generate coexpression networks in T1DM and T2DM. Functional pathways in highly correlated modules of T1DM were enriched by gene set enrichment analysis (GSEA). We next filtered the differentially expressed genes (DEGs) and revealed their dynamic expression profiles in T1DM with or without insulin treatment. Furthermore, dynamic clusters and dynamic protein-protein interaction networks were identified. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis was developed in dynamic clusters. RESULTS WGCNA disclosed 12 distinct gene modules, and distinguished between correlated networks in T1DM and T2DM. Two modules were closely associated with T1DM. GSEA showed that the immune response and response to cytokines were enriched in the T1DM highly correlated module. Next, we screened 44 DEGs in newly diagnosed T1DM compared with healthy donors, and 71 DEGs in 1-month and 97 DEGs in 4-month insulin treatment groups compared with newly diagnosed T1DM. Dynamic expression profiles of DEGs indicated the potential targets for T1DM treatment. Moreover, four molecular dynamic clusters were analyzed in newly diagnosed and insulin-treated T1DM. Functional annotation showed that these clusters were mainly enriched in the IL-17 signaling pathway, nuclear factor-ϰB signaling pathway, and tumor necrosis factor signaling pathway. CONCLUSION The results indicate potential drug targets or clinical efficacy markers, as well as demonstrating the underlying molecular mechanisms of T1DM treatment.
Collapse
Affiliation(s)
- Lu Li
- Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
- Correspondence: Lu Li Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, Zhejiang, People’s Republic of ChinaTel +865 718 723 6675Fax +865 718 723 6675Email
| | - Zongfu Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Xi Yang
- Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
171
|
Hoffman GE, Schrode N, Flaherty E, Brennand KJ. New considerations for hiPSC-based models of neuropsychiatric disorders. Mol Psychiatry 2019; 24:49-66. [PMID: 29483625 PMCID: PMC6109625 DOI: 10.1038/s41380-018-0029-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
The development of human-induced pluripotent stem cells (hiPSCs) has made possible patient-specific modeling across the spectrum of human disease. Here, we discuss recent advances in psychiatric genomics and post-mortem studies that provide critical insights concerning cell-type composition and sample size that should be considered when designing hiPSC-based studies of complex genetic disease. We review recent hiPSC-based models of SZ, in light of our new understanding of critical power limitations in the design of hiPSC-based studies of complex genetic disorders. Three possible solutions are a movement towards genetically stratified cohorts of rare variant patients, application of CRISPR technologies to engineer isogenic neural cells to study the impact of common variants, and integration of advanced genetics and hiPSC-based datasets in future studies. Overall, we emphasize that to advance the reproducibility and relevance of hiPSC-based studies, stem cell biologists must contemplate statistical and biological considerations that are already well accepted in the field of genetics. We conclude with a discussion of the hypothesis of biological convergence of disease-through molecular, cellular, circuit, and patient level phenotypes-and how this might emerge through hiPSC-based studies.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nadine Schrode
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erin Flaherty
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristen J Brennand
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
172
|
Erfanian Omidvar M, Ghaedi H, Kazerouni F, Kalbasi S, Shanaki M, Miraalamy G, Zare A, Rahimipour A. Clinical significance of long noncoding RNA VIM-AS1 and CTBP1-AS2 expression in type 2 diabetes. J Cell Biochem 2018; 120:9315-9323. [PMID: 30506719 DOI: 10.1002/jcb.28206] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIMS The risk of type 2 diabetes (T2D) is determined by a combination of genetic and environmental factors. Multiple studies have proposed that long noncoding RNAs (lncRNAs) are crucial molecules in regulating several biological processes and complex diseases. The study was aimed at investigating the association between the expression levels of lncRNA VIM-AS1, lncRNA CTBP1-AS2, and T2D susceptibility. METHODS lncRNA VIM-AS1 and lncRNA CTBP1-AS2 in the peripheral blood mononuclear cell (PBMC) of 100 healthy individuals and 100 T2D patients were collected for Quantitative Real-Time RT-PCR analysis. A logistic regression was performed to understand whether the likelihood of T2D can be predicted based on the expression levels of lncRNA VIM-AS1 and lncRNA CTBP1-AS2. Receiver operating characteristic (ROC) analysis was also performed to determine the statistical analysis of VIM-AS1 and CTBP1-AS2 levels in 200 samples. RESULTS Our results display that decreased levels of VIM-AS1 and CTBP1-AS2 in PBMC were associated with diabetes in Iranian population. The logistic regression revealed that Systolic blood pressure (SBP), low-density lipoprotein cholesterol (LDL-C), Fasting blood glucose (FBG) and CTBP1-AS2 are substantial predictors of T2D. The ROC analysis of CTBP1-AS2 revealed the area under the ROC curve (AUC) of 0.68 with a sensitivity of 58.7% and specificity of 75.3% in distinguishing nondiabetic from diabetic subjects. The ROC analysis of VIM-AS1 determined AUC of 0.63 with a sensitivity of 56.1% and specificity of 68.37% in distinguishing the two diagnostic groups. CONCLUSION lncRNA VIM-AS1 and lncRNA CTBP1-AS2 expression levels are associated with T2D susceptibility.
Collapse
Affiliation(s)
- Maryam Erfanian Omidvar
- Department of Medical Laboratory Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faranak Kazerouni
- Department of Medical Laboratory Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Kalbasi
- Department of endocrinology, Loghman Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shanaki
- Department of Medical Laboratory Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Miraalamy
- Ali-Asghar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zare
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Rahimipour
- Department of Medical Laboratory Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
173
|
Nammo T, Udagawa H, Funahashi N, Kawaguchi M, Uebanso T, Hiramoto M, Nishimura W, Yasuda K. Genome-wide profiling of histone H3K27 acetylation featured fatty acid signalling in pancreatic beta cells in diet-induced obesity in mice. Diabetologia 2018; 61:2608-2620. [PMID: 30284014 DOI: 10.1007/s00125-018-4735-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/17/2018] [Indexed: 01/31/2023]
Abstract
AIMS/HYPOTHESIS Epigenetic regulation of gene expression has been implicated in the pathogenesis of obesity and type 2 diabetes. However, detailed information, such as key transcription factors in pancreatic beta cells that mediate environmental effects, is not yet available. METHODS To analyse genome-wide cis-regulatory profiles and transcriptome of pancreatic islets derived from a diet-induced obesity (DIO) mouse model, we conducted chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-Seq) of histone H3 lysine 27 acetylation (histone H3K27ac) and high-throughput RNA sequencing. Transcription factor-binding motifs enriched in differential H3K27ac regions were examined by de novo motif analysis. For the predicted transcription factors, loss of function experiments were performed by transfecting specific siRNA in INS-1, a rat beta cell line, with and without palmitate treatment. Epigenomic and transcriptional changes of possible target genes were evaluated by ChIP and quantitative RT-PCR. RESULTS After long-term feeding with a high-fat diet, C57BL/6J mice were obese and mildly glucose intolerant. Among 39,350 islet cis-regulatory regions, 13,369 and 4610 elements showed increase and decrease in ChIP-Seq signals, respectively, significantly associated with global change in gene expression. Remarkably, increased H3K27ac showed a distinctive genomic localisation, mainly in the proximal-promoter regions, revealing enriched elements for nuclear respiratory factor 1 (NRF1), GA repeat binding protein α (GABPA) and myocyte enhancer factor 2A (MEF2A) by de novo motif analysis, whereas decreased H3K27ac was enriched for v-maf musculoaponeurotic fibrosarcoma oncogene family protein K (MAFK), a known negative regulator of beta cells. By siRNA-mediated knockdown of NRF1, GABPA or MEF2A we found that INS-1 cells exhibited downregulation of fatty acid β-oxidation genes in parallel with decrease in the associated H3K27ac. Furthermore, in line with the epigenome in DIO mice, palmitate treatment caused increase in H3K27ac and induction of β-oxidation genes; these responses were blunted when NRF1, GABPA or MEF2A were suppressed. CONCLUSIONS/INTERPRETATION These results suggest novel roles for DNA-binding proteins and fatty acid signalling in obesity-induced epigenomic regulation of beta cell function. DATA AVAILABILITY The next-generation sequencing data in the present study were deposited at ArrayExpress. RNA-Seq: Dataset name: ERR2538129 (Control), ERR2538130 (Diet-induced obesity) Repository name and number: E-MTAB-6718 - RNA-Seq of pancreatic islets derived from mice fed a long-term high-fat diet against chow-fed controls. ChIP-Seq: Dataset name: ERR2538131 (Control), ERR2538132 (Diet-induced obesity) Repository name and number: E-MTAB-6719 - H3K27ac ChIP-Seq of pancreatic islets derived from mice fed a long-term high-fat diet (HFD) against chow-fed controls.
Collapse
Affiliation(s)
- Takao Nammo
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.
| | - Haruhide Udagawa
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Nobuaki Funahashi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Miho Kawaguchi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masaki Hiramoto
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Wataru Nishimura
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
- Department of Molecular Biology, International University of Health and Welfare School of Medicine, Narita, Chiba, Japan
- Division of Anatomy, Bio-imaging and Neuro-cell Science, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.
| |
Collapse
|
174
|
Zhang Q, Liu HM, Lv WQ, He JY, Xia X, Zhang WD, Deng HW, Sun CQ. Additional common variants associated with type 2 diabetes and coronary artery disease detected using a pleiotropic cFDR method. J Diabetes Complications 2018; 32:1105-1112. [PMID: 30270018 PMCID: PMC6743331 DOI: 10.1016/j.jdiacomp.2018.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/11/2018] [Accepted: 09/02/2018] [Indexed: 12/27/2022]
Abstract
Genome-wide association studies (GWASs) have been performed extensively in diverse populations to identify single nucleotide polymorphisms (SNPs) associated with complex diseases or traits. However, to date, the SNPs identified fail to explain a large proportion of the variance of the traits/diseases. GWASs on type 2 diabetes (T2D) and coronary artery disease (CARD) are generally performed as single-trait studies, rather than analyzing the related traits simultaneously. Despite the extensive evidence suggesting that these two phenotypes share both genetic and environmental risk factors, the shared overlapping genetic biological mechanisms between these traits remain largely unexplored. Here, we adopted a recently developed genetic pleiotropic conditional false discovery rate (cFDR) approach to discover novel loci associated with T2D and CARD by incorporating the summary statistics from existing GWASs of these two traits. Applying the cFDR level of 0.05, 33 loci were identified for T2D and 34 loci for CARD, 9 of which for both. By incorporating pleiotropic effects into a conditional analysis framework, we observed that there is significant pleiotropic enrichment between T2D and CARD. These findings may provide novel insights into the etiology of T2D and CARD, as well as the processes that may influence disease development both individually and jointly.
Collapse
Affiliation(s)
- Qiang Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Hui-Min Liu
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Wan-Qiang Lv
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Jing-Yang He
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Xin Xia
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Wei-Dong Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China
| | - Hong-Wen Deng
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China; Center for Bioinformatics and Genomics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Chang-Qing Sun
- College of Public Health, Zhengzhou University, Zhengzhou, No. 100 Kexue Road, High-Tech Development Zone Of States, PR China.
| |
Collapse
|
175
|
Firneisz G, Rosta K, Al-Aissa Z, Hadarits O, Harreiter J, Nádasdi Á, Bancher-Todesca D, Németh L, Igaz P, Rigó J, Sziller I, Kautzky-Willer A, Somogyi A. The MTNR1B rs10830963 Variant in Interaction with Pre-Pregnancy BMI is a Pharmacogenetic Marker for the Initiation of Antenatal Insulin Therapy in Gestational Diabetes Mellitus. Int J Mol Sci 2018; 19:E3734. [PMID: 30477160 PMCID: PMC6321391 DOI: 10.3390/ijms19123734] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 11/26/2022] Open
Abstract
The rs10830963 variant of the Melatonin Receptor 1B (MTNR1B) gene is associated with the development of gestational diabetes mellitus (GDM). We hypothesized that carrying the rs10830963/G risk allele had effect on antenatal insulin therapy (AIT) initiation in GDM in a body mass index (BMI)-dependent manner. Design: In this post hoc analysis the MTNR1B rs10830963 genotype and the clinical data of 211 Caucasian GDM patients were assessed. As a first step, a pre-pregnancy BMI threshold was determined where the effect of MTNR1B rs10830963/G allele carrying on AIT initiation was the most significant using logistic regression. Maternal age adjusted real-life odds ratios (OR) values were calculated. The chi-square test was also used to calculate the p value and 10.000 bootstrap simulations were performed in each case to re-assess the statistical power and the OR. Carrying the MTNR1B rs10830963/G allele increased the odds of AIT initiation (OR = 5.2, p = 0.02 [χ² test], statistical power = 0.53) in GDM patients with pre-pregnancy BMI ≥ 29 kg/m². The statistical power reached 0.77, when the pre-pregnancy BMI cutoff of 27 kg/m² was used and the genetic effect on AIT initiation was still significant, but only using the logistic regression model. Carrying the MTNR1B rs10830963/G risk allele-in interaction with pre-pregnancy BMI-is likely be considered as a candidate pharmacogenetic marker of antenatal insulin therapy initiation and should be further assessed in precision medicine trials in GDM.
Collapse
Affiliation(s)
- Gábor Firneisz
- 2nd Department of Internal Medicine, Semmelweis University, H-1088 Budapest, Hungary.
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences⁻Semmelweis University, H-1088 Budapest, Hungary.
| | - Klara Rosta
- Department of Obstetrics and Gynecology, Medical University of Vienna, A-1090 Vienna, Austria.
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary.
| | - Zahra Al-Aissa
- 2nd Department of Internal Medicine, Semmelweis University, H-1088 Budapest, Hungary.
| | - Orsolya Hadarits
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary.
| | - Jürgen Harreiter
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Ákos Nádasdi
- 2nd Department of Internal Medicine, Semmelweis University, H-1088 Budapest, Hungary.
| | - Dagmar Bancher-Todesca
- Department of Obstetrics and Gynecology, Medical University of Vienna, A-1090 Vienna, Austria.
| | - László Németh
- Department of Probability Theory and Statistics, Eötvös Loránd University, H-1088 Budapest, Hungary.
| | - Péter Igaz
- 2nd Department of Internal Medicine, Semmelweis University, H-1088 Budapest, Hungary.
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences⁻Semmelweis University, H-1088 Budapest, Hungary.
| | - János Rigó
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary.
| | - István Sziller
- Department of Obstetrics and Gynecology, Szent Imre Teaching Hospital, H-1088 Budapest, Hungary.
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Anikó Somogyi
- 2nd Department of Internal Medicine, Semmelweis University, H-1088 Budapest, Hungary.
| |
Collapse
|
176
|
Mahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Steinthorsdottir V, Scott RA, Grarup N, Cook JP, Schmidt EM, Wuttke M, Sarnowski C, Mägi R, Nano J, Gieger C, Trompet S, Lecoeur C, Preuss M, Prins BP, Guo X, Bielak LF, DIAMANTE consortium, Bennett AJ, Bork-Jensen J, Brummett CM, Canouil M, Eckardt KU, Fischer K, Kardia SLR, Kronenberg F, Läll K, Liu CT, Locke AE, Luan J, Ntalla I, Nylander V, Schönherr S, Schurmann C, Yengo L, Bottinger EP, Brandslund I, Christensen C, Dedoussis G, Florez JC, ford I, Franco OH, Frayling TM, Giedraitis V, Hackinger S, Hattersley AT, Herder C, Ikram MA, Ingelsson M, Jørgensen ME, Jørgensen T, Kriebel J, Kuusisto J, Ligthart S, Lindgren CM, Linneberg A, Lyssenko V, Mamakou V, Meitinger T, Mohlke KL, Morris AD, Nadkarni G, Pankow JS, Peters A, Sattar N, Stančáková A, Strauch K, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tuomilehto J, Witte DR, Dupuis J, Peyser PA, Zeggini E, Loos RJF, Froguel P, Ingelsson E, Lind L, Groop L, Laakso M, Collins FS, Jukema JW, Palmer CNA, Grallert H, Metspalu A, Dehghan A, Köttgen A, Abecasis G, Meigs JB, Rotter JI, Marchini J, Pedersen O, Hansen T, et alMahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Steinthorsdottir V, Scott RA, Grarup N, Cook JP, Schmidt EM, Wuttke M, Sarnowski C, Mägi R, Nano J, Gieger C, Trompet S, Lecoeur C, Preuss M, Prins BP, Guo X, Bielak LF, DIAMANTE consortium, Bennett AJ, Bork-Jensen J, Brummett CM, Canouil M, Eckardt KU, Fischer K, Kardia SLR, Kronenberg F, Läll K, Liu CT, Locke AE, Luan J, Ntalla I, Nylander V, Schönherr S, Schurmann C, Yengo L, Bottinger EP, Brandslund I, Christensen C, Dedoussis G, Florez JC, ford I, Franco OH, Frayling TM, Giedraitis V, Hackinger S, Hattersley AT, Herder C, Ikram MA, Ingelsson M, Jørgensen ME, Jørgensen T, Kriebel J, Kuusisto J, Ligthart S, Lindgren CM, Linneberg A, Lyssenko V, Mamakou V, Meitinger T, Mohlke KL, Morris AD, Nadkarni G, Pankow JS, Peters A, Sattar N, Stančáková A, Strauch K, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tuomilehto J, Witte DR, Dupuis J, Peyser PA, Zeggini E, Loos RJF, Froguel P, Ingelsson E, Lind L, Groop L, Laakso M, Collins FS, Jukema JW, Palmer CNA, Grallert H, Metspalu A, Dehghan A, Köttgen A, Abecasis G, Meigs JB, Rotter JI, Marchini J, Pedersen O, Hansen T, Langenberg C, Wareham NJ, Stefansson K, Gloyn AL, Morris AP, Boehnke M, McCarthy MI. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat Genet 2018; 50:1505-1513. [PMID: 30297969 PMCID: PMC6287706 DOI: 10.1038/s41588-018-0241-6] [Show More Authors] [Citation(s) in RCA: 1197] [Impact Index Per Article: 171.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 08/10/2018] [Indexed: 12/30/2022]
Abstract
We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence).
Collapse
Affiliation(s)
- Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Daniel Taliun
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Thurner
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Neil R Robertson
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Jason M Torres
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - N William Rayner
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | | | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, L69 3GA, UK
| | - Ellen M Schmidt
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Chloé Sarnowski
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Jana Nano
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology 2, Helmholtz Zentrum München, German Research Center for Environmental Health, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Cécile Lecoeur
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Bram Peter Prins
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | - Amanda J Bennett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Jette Bork-Jensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, 48109, US
| | - Mickaël Canouil
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care Charité, University Medicine Berlin, Berlin, 10117, Germany and German Chronic Kidney Disease study
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Sharon LR Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, 6020, Austria and German Chronic Kidney Disease study
| | - Kristi Läll
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
- Institute of Mathematical Statistics, University of Tartu, Tartu, Estonia
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
| | - Adam E Locke
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Division of Genomics & Bioinformatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vibe Nylander
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Sebastian Schönherr
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, 6020, Austria and German Chronic Kidney Disease study
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Loïc Yengo
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Ivan Brandslund
- Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark
- Department of Clinical Biochemistry, Vejle Hospital, Vejle, 7100, Denmark
| | | | - George Dedoussis
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, 17671, Greece
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA, 02142, USA
| | - Ian ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, EX1 2LU, UK
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Sophie Hackinger
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Andrew T Hattersley
- University of Exeter Medical School, University of Exeter, Exeter, EX2 5DW, UK
| | - Christian Herder
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Marit E Jørgensen
- Steno Diabetes Center Copenhagen, Gentofte, 2820, Denmark
- National Institute of Public Health, Southern Denmark University, Copenhagen, 1353, Denmark
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup, 2600, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Symen Ligthart
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, 02142, USA
- Big Data Institute, Li Ka Shing Centre For Health Information and Discovery, University of Oxford, Oxford, OX37BN, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup, 2600, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, 20502, Sweden
| | - Vasiliki Mamakou
- Dromokaiteio Psychiatric Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Munich, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, 27599, USA
| | - Andrew D Morris
- Clinical Research Centre, Centre for Molecular Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
- The Usher Institute to the Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10069, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, US
| | - Annette Peters
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, 81675, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, 80802, Germany
| | - Kent D Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Barbara Thorand
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Jaakko Tuomilehto
- Department of Health, National Institute for Health and Welfare, Helsinki, 00271, Finland
- Dasman Diabetes Institute, Dasman, 15462, Kuwait
- Department of Neuroscience and Preventive Medicine, Danube-University Krems, Krems, 3500, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, 01702, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Eleftheria Zeggini
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
- Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Philippe Froguel
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, W12 0NN, UK
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, US
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, 75185, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, 20502, Sweden
- Finnish Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Francis S Collins
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Clinical Cooparation Group Type 2 Diabetes, Helmholtz Zentrum München, Ludwig-Maximillians University Munich, Germany
- Clinical Cooparation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Technical University Munich, Germany
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Goncalo Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - James B Meigs
- General Medicine Division, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Jerome I Rotter
- Departments of Pediatrics and Medicine, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Jonathan Marchini
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX1 3TG, UK
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, 5000, Denmark
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Anna L Gloyn
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool, L69 3GA, UK
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| |
Collapse
|
177
|
Khetan S, Kursawe R, Youn A, Lawlor N, Jillette A, Marquez EJ, Ucar D, Stitzel ML. Type 2 Diabetes-Associated Genetic Variants Regulate Chromatin Accessibility in Human Islets. Diabetes 2018; 67:2466-2477. [PMID: 30181159 PMCID: PMC6198349 DOI: 10.2337/db18-0393] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/22/2018] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes (T2D) is a complex disorder in which both genetic and environmental risk factors contribute to islet dysfunction and failure. Genome-wide association studies (GWAS) have linked single nucleotide polymorphisms (SNPs), most of which are noncoding, in >200 loci to islet dysfunction and T2D. Identification of the putative causal variants and their target genes and whether they lead to gain or loss of function remains challenging. Here, we profiled chromatin accessibility in pancreatic islet samples from 19 genotyped individuals and identified 2,949 SNPs associated with in vivo cis-regulatory element use (i.e., chromatin accessibility quantitative trait loci [caQTL]). Among the caQTLs tested (n = 13) using luciferase reporter assays in MIN6 β-cells, more than half exhibited effects on enhancer activity that were consistent with in vivo chromatin accessibility changes. Importantly, islet caQTL analysis nominated putative causal SNPs in 13 T2D-associated GWAS loci, linking 7 and 6 T2D risk alleles, respectively, to gain or loss of in vivo chromatin accessibility. By investigating the effect of genetic variants on chromatin accessibility in islets, this study is an important step forward in translating T2D-associated GWAS SNP into functional molecular consequences.
Collapse
Affiliation(s)
- Shubham Khetan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Ahrim Youn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Nathan Lawlor
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | | | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT
- Institute of Systems Genomics, University of Connecticut, Farmington, CT
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT
- Institute of Systems Genomics, University of Connecticut, Farmington, CT
| |
Collapse
|
178
|
Lipid-associated genetic polymorphisms are associated with FBP and LDL-c levels among myocardial infarction patients in Chinese population. Gene 2018; 676:22-28. [DOI: 10.1016/j.gene.2018.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/29/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022]
|
179
|
Kim YK, Nam KI, Song J. The Glymphatic System in Diabetes-Induced Dementia. Front Neurol 2018; 9:867. [PMID: 30429819 PMCID: PMC6220044 DOI: 10.3389/fneur.2018.00867] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
The glymphatic system has emerged as an important player in central nervous system (CNS) diseases, by regulating the vasculature impairment, effectively controlling the clearance of toxic peptides, modulating activity of astrocytes, and being involved in the circulation of neurotransmitters in the brain. Recently, several studies have indicated decreased activity of the glymphatic pathway under diabetes conditions such as in insulin resistance and hyperglycemia. Furthermore, diabetes leads to the disruption of the blood-brain barrier and decrease of apolipoprotein E (APOE) expression and the secretion of norepinephrine in the brain, involving the impairment of the glymphatic pathway and ultimately resulting in cognitive decline. Considering the increased prevalence of diabetes-induced dementia worldwide, the relationship between the glymphatic pathway and diabetes-induced dementia should be investigated and the mechanisms underlying their relationship should be discussed to promote the development of an effective therapeutic approach in the near future. Here, we have reviewed recent evidence for the relationship between glymphatic pathway dysfunction and diabetes. We highlight that the enhancement of the glymphatic system function during sleep may be beneficial to the attenuation of neuropathology in diabetes-induced dementia. Moreover, we suggest that improving glymphatic system activity may be a potential therapeutic strategy for the prevention of diabetes-induced dementia.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, South Korea.,Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| | - Kwang Il Nam
- Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| | - Juhyun Song
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea.,Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
180
|
Strohmaier S, Devore EE, Zhang Y, Schernhammer ES. A Review of Data of Findings on Night Shift Work and the Development of DM and CVD Events: a Synthesis of the Proposed Molecular Mechanisms. Curr Diab Rep 2018; 18:132. [PMID: 30343445 PMCID: PMC6209035 DOI: 10.1007/s11892-018-1102-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Night shift work has become highly prevalent in our 24/7 societies, with up to 18% of the US work force working alternate shift schedules. However, studies indicate that there may be adverse health effects of chronic night work across diverse populations. These effects are likely due to misalignment of the circadian system with work schedules, mediated by the system's primary marker melatonin as well as other downstream molecules. RECENT FINDINGS Melatonin has multiple biologic actions that are relevant to cardiometabolic disease, including modulation of oxidative stress, inflammation, and (via the melatonin receptor) vasoconstriction. Behavioral traits, such as chronotype and meal timing, have recently been shown to interact with the effects of night work on cardiometabolic health. Together with recent findings suggesting a role for circadian genes in cardiometabolic risk, the interactions of night shift work and behavioral traits are likely to facilitate novel treatment and prevention approaches for cardiovascular disease and type 2 diabetes, incorporating aspects of clock and timing.
Collapse
Affiliation(s)
- S. Strohmaier
- 0000 0000 9259 8492grid.22937.3dDepartment of Epidemiology, Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - E. E. Devore
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - Y. Zhang
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - E. S. Schernhammer
- 0000 0000 9259 8492grid.22937.3dDepartment of Epidemiology, Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
- 000000041936754Xgrid.38142.3cDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| |
Collapse
|
181
|
Fernández-Rhodes L, Howard AG, Graff M, Isasi CR, Highland HM, Young KL, Parra E, Below JE, Qi Q, Kaplan RC, Justice AE, Papanicolaou G, Laurie CC, Grant SFA, Haiman C, Loos RJF, North KE. Complex patterns of direct and indirect association between the transcription Factor-7 like 2 gene, body mass index and type 2 diabetes diagnosis in adulthood in the Hispanic Community Health Study/Study of Latinos. BMC OBESITY 2018; 5:26. [PMID: 30305909 PMCID: PMC6167893 DOI: 10.1186/s40608-018-0200-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/23/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Genome-wide association studies have implicated the transcription factor 7-like 2 (TCF7L2) gene in type 2 diabetes risk, and more recently, in decreased body mass index. Given the contrary direction of genetic effects on these two traits, it has been suggested that the observed association with body mass index may reflect either selection bias or a complex underlying biology at TCF7L2. METHODS Using 9031 Hispanic/Latino adults (21-76 years) with complete weight history and genetic data from the community-based Hispanic Community Health Study/Study of Latinos (HCHS/SOL, Baseline 2008-2011), we estimated the multivariable association between the additive number of type 2 diabetes increasing-alleles at TCF7L2 (rs7903146-T) and body mass index. We then used structural equation models to simultaneously model the genetic association on changes in body mass index across the life course and estimate the odds of type 2 diabetes per TCF7L2 risk allele. RESULTS We observed both significant increases in type 2 diabetes prevalence at examination (independent of body mass index) and decreases in mean body mass index and waist circumference across genotypes at rs7903146. We observed a significant multivariable association between the additive number of type 2 diabetes-risk alleles and lower body mass index at examination. In our structured modeling, we observed non-significant inverse direct associations between rs7903146-T and body mass index at ages 21 and 45 years, and a significant positive association between rs7903146-T and type 2 diabetes onset in both middle and late adulthood. CONCLUSIONS Herein, we replicated the protective effect of rs7930146-T on body mass index at multiple time points in the life course, and observed that these effects were not explained by past type 2 diabetes status in our structured modeling. The robust replication of the negative effects of TCF7L2 on body mass index in multiple samples, including in our diverse Hispanic/Latino community-based sample, supports a growing body of literature on the complex biologic mechanism underlying the functional consequences of TCF7L2 on obesity and type 2 diabetes across the life course.
Collapse
Affiliation(s)
- Lindsay Fernández-Rhodes
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
- Carolina Population Center, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
| | - Annie Green Howard
- Carolina Population Center, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Mariaelisa Graff
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
| | - Carmen R. Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY USA
| | - Heather M. Highland
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
| | - Kristin L. Young
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
| | - Esteban Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON Canada
| | - Jennifer E. Below
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY USA
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY USA
| | - Anne E. Justice
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - George Papanicolaou
- Epidemiology Branch, National Heart Lung and Blood Institute, Bethesda, MD USA
| | - Cathy C. Laurie
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA USA
| | - Struan F. A. Grant
- Divisions of Human Genetics and Endocrinology, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA USA
| | - Christopher Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Ruth J. F. Loos
- Charles R. Bronfman Instituted for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Kari E. North
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 123 W Franklin St, Building C, Chapel Hill, NC USA
| |
Collapse
|
182
|
Fine-mapping and functional studies highlight potential causal variants for rheumatoid arthritis and type 1 diabetes. Nat Genet 2018; 50:1366-1374. [PMID: 30224649 DOI: 10.1038/s41588-018-0216-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/30/2018] [Indexed: 12/19/2022]
Abstract
To define potentially causal variants for autoimmune disease, we fine-mapped1,2 76 rheumatoid arthritis (11,475 cases, 15,870 controls)3 and type 1 diabetes loci (9,334 cases, 11,111 controls)4. After sequencing 799 1-kilobase regulatory (H3K4me3) regions within these loci in 568 individuals, we observed accurate imputation for 89% of common variants. We defined credible sets of ≤5 causal variants at 5 rheumatoid arthritis and 10 type 1 diabetes loci. We identified potentially causal missense variants at DNASE1L3, PTPN22, SH2B3, and TYK2, and noncoding variants at MEG3, CD28-CTLA4, and IL2RA. We also identified potential candidate causal variants at SIRPG and TNFAIP3. Using functional assays, we confirmed allele-specific protein binding and differential enhancer activity for three variants: the CD28-CTLA4 rs117701653 SNP, MEG3 rs34552516 indel, and TNFAIP3 rs35926684 indel.
Collapse
|
183
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
184
|
Timmons JA, Atherton PJ, Larsson O, Sood S, Blokhin IO, Brogan RJ, Volmar CH, Josse AR, Slentz C, Wahlestedt C, Phillips SM, Phillips BE, Gallagher IJ, Kraus WE. A coding and non-coding transcriptomic perspective on the genomics of human metabolic disease. Nucleic Acids Res 2018; 46:7772-7792. [PMID: 29986096 PMCID: PMC6125682 DOI: 10.1093/nar/gky570] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 05/23/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies (GWAS), relying on hundreds of thousands of individuals, have revealed >200 genomic loci linked to metabolic disease (MD). Loss of insulin sensitivity (IS) is a key component of MD and we hypothesized that discovery of a robust IS transcriptome would help reveal the underlying genomic structure of MD. Using 1,012 human skeletal muscle samples, detailed physiology and a tissue-optimized approach for the quantification of coding (>18,000) and non-coding (>15,000) RNA (ncRNA), we identified 332 fasting IS-related genes (CORE-IS). Over 200 had a proven role in the biochemistry of insulin and/or metabolism or were located at GWAS MD loci. Over 50% of the CORE-IS genes responded to clinical treatment; 16 quantitatively tracking changes in IS across four independent studies (P = 0.0000053: negatively: AGL, G0S2, KPNA2, PGM2, RND3 and TSPAN9 and positively: ALDH6A1, DHTKD1, ECHDC3, MCCC1, OARD1, PCYT2, PRRX1, SGCG, SLC43A1 and SMIM8). A network of ncRNA positively related to IS and interacted with RNA coding for viral response proteins (P < 1 × 10-48), while reduced amino acid catabolic gene expression occurred without a change in expression of oxidative-phosphorylation genes. We illustrate that combining in-depth physiological phenotyping with robust RNA profiling methods, identifies molecular networks which are highly consistent with the genetics and biochemistry of human metabolic disease.
Collapse
Affiliation(s)
- James A Timmons
- Division of Genetics and Molecular Medicine, King's College London, London, UK
- Scion House, Stirling University Innovation Park, Stirling, UK
| | | | - Ola Larsson
- Department of Oncology-Pathology, Science For Life Laboratory, Stockholm, Sweden
| | - Sanjana Sood
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | | | - Robert J Brogan
- Scion House, Stirling University Innovation Park, Stirling, UK
| | | | | | - Cris Slentz
- Duke University School of Medicine, Durham, USA
| | - Claes Wahlestedt
- Department of Oncology-Pathology, Science For Life Laboratory, Stockholm, Sweden
| | | | | | - Iain J Gallagher
- Scion House, Stirling University Innovation Park, Stirling, UK
- School of Health Sciences and Sport, University of Stirling, Stirling, UK
| | | |
Collapse
|
185
|
Locke JM, Saint-Martin C, Laver TW, Patel KA, Wood AR, Sharp SA, Ellard S, Bellanné-Chantelot C, Hattersley AT, Harries LW, Weedon MN. The Common HNF1A Variant I27L Is a Modifier of Age at Diabetes Diagnosis in Individuals With HNF1A-MODY. Diabetes 2018; 67:1903-1907. [PMID: 29895593 PMCID: PMC6109380 DOI: 10.2337/db18-0133] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/05/2018] [Indexed: 01/03/2023]
Abstract
There is wide variation in the age at diagnosis of diabetes in individuals with maturity-onset diabetes of the young (MODY) due to a mutation in the HNF1A gene. We hypothesized that common variants at the HNF1A locus (rs1169288 [I27L], rs1800574 [A98V]), which are associated with type 2 diabetes susceptibility, may modify age at diabetes diagnosis in individuals with HNF1A-MODY. Meta-analysis of two independent cohorts, comprising 781 individuals with HNF1A-MODY, found no significant associations between genotype and age at diagnosis. However after stratifying according to type of mutation (protein-truncating variant [PTV] or missense), we found each 27L allele to be associated with a 1.6-year decrease (95% CI -2.6, -0.7) in age at diagnosis, specifically in the subset (n = 444) of individuals with a PTV. The effect size was similar and significant across the two independent cohorts of individuals with HNF1A-MODY. We report a robust genetic modifier of HNF1A-MODY age at diagnosis that further illustrates the strong effect of genetic variation within HNF1A upon diabetes phenotype.
Collapse
Affiliation(s)
- Jonathan M Locke
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K.
| | - Cécile Saint-Martin
- Department of Genetics, Pitié-Salpétrière Hospital, Assistance Publique-Hôpitaux de Paris, and Pierre et Marie Curie University, Paris, France
| | - Thomas W Laver
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Andrew R Wood
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Seth A Sharp
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Christine Bellanné-Chantelot
- Department of Genetics, Pitié-Salpétrière Hospital, Assistance Publique-Hôpitaux de Paris, and Pierre et Marie Curie University, Paris, France
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Lorna W Harries
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| | - Michael N Weedon
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, U.K
| |
Collapse
|
186
|
Udler MS, Kim J, von Grotthuss M, Bonàs-Guarch S, Cole JB, Chiou J, Christopher D. Anderson on behalf of METASTROKE and the ISGC, Boehnke M, Laakso M, Atzmon G, Glaser B, Mercader JM, Gaulton K, Flannick J, Getz G, Florez JC. Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A soft clustering analysis. PLoS Med 2018; 15:e1002654. [PMID: 30240442 PMCID: PMC6150463 DOI: 10.1371/journal.pmed.1002654] [Citation(s) in RCA: 347] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a heterogeneous disease for which (1) disease-causing pathways are incompletely understood and (2) subclassification may improve patient management. Unlike other biomarkers, germline genetic markers do not change with disease progression or treatment. In this paper, we test whether a germline genetic approach informed by physiology can be used to deconstruct T2D heterogeneity. First, we aimed to categorize genetic loci into groups representing likely disease mechanistic pathways. Second, we asked whether the novel clusters of genetic loci we identified have any broad clinical consequence, as assessed in four separate subsets of individuals with T2D. METHODS AND FINDINGS In an effort to identify mechanistic pathways driven by established T2D genetic loci, we applied Bayesian nonnegative matrix factorization (bNMF) clustering to genome-wide association study (GWAS) results for 94 independent T2D genetic variants and 47 diabetes-related traits. We identified five robust clusters of T2D loci and traits, each with distinct tissue-specific enhancer enrichment based on analysis of epigenomic data from 28 cell types. Two clusters contained variant-trait associations indicative of reduced beta cell function, differing from each other by high versus low proinsulin levels. The three other clusters displayed features of insulin resistance: obesity mediated (high body mass index [BMI] and waist circumference [WC]), "lipodystrophy-like" fat distribution (low BMI, adiponectin, and high-density lipoprotein [HDL] cholesterol, and high triglycerides), and disrupted liver lipid metabolism (low triglycerides). Increased cluster genetic risk scores were associated with distinct clinical outcomes, including increased blood pressure, coronary artery disease (CAD), and stroke. We evaluated the potential for clinical impact of these clusters in four studies containing individuals with T2D (Metabolic Syndrome in Men Study [METSIM], N = 487; Ashkenazi, N = 509; Partners Biobank, N = 2,065; UK Biobank [UKBB], N = 14,813). Individuals with T2D in the top genetic risk score decile for each cluster reproducibly exhibited the predicted cluster-associated phenotypes, with approximately 30% of all individuals assigned to just one cluster top decile. Limitations of this study include that the genetic variants used in the cluster analysis were restricted to those associated with T2D in populations of European ancestry. CONCLUSION Our approach identifies salient T2D genetically anchored and physiologically informed pathways, and supports the use of genetics to deconstruct T2D heterogeneity. Classification of patients by these genetic pathways may offer a step toward genetically informed T2D patient management.
Collapse
Affiliation(s)
- Miriam S. Udler
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jaegil Kim
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Marcin von Grotthuss
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Sílvia Bonàs-Guarch
- Barcelona Supercomputing Center (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - Joanne B. Cole
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Joshua Chiou
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
| | | | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Gil Atzmon
- Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Department of Medicine; Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Genetics, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Josep M. Mercader
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Barcelona Supercomputing Center (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - Kyle Gaulton
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
| | - Jason Flannick
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Department of Genetics, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Jose C. Florez
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
187
|
Karamitri A, Plouffe B, Bonnefond A, Chen M, Gallion J, Guillaume JL, Hegron A, Boissel M, Canouil M, Langenberg C, Wareham NJ, Le Gouill C, Lukasheva V, Lichtarge O, Froguel P, Bouvier M, Jockers R. Type 2 diabetes-associated variants of the MT 2 melatonin receptor affect distinct modes of signaling. Sci Signal 2018; 11:11/545/eaan6622. [PMID: 30154102 DOI: 10.1126/scisignal.aan6622] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melatonin is produced during the night and regulates sleep and circadian rhythms. Loss-of-function variants in MTNR1B, which encodes the melatonin receptor MT2, a G protein-coupled receptor (GPCR), are associated with an increased risk of type 2 diabetes (T2D). To identify specific T2D-associated signaling pathway(s), we profiled the signaling output of 40 MT2 variants by monitoring spontaneous (ligand-independent) and melatonin-induced activation of multiple signaling effectors. Genetic association analysis showed that defects in the melatonin-induced activation of Gαi1 and Gαz proteins and in spontaneous β-arrestin2 recruitment to MT2 were the most statistically significantly associated with an increased T2D risk. Computational variant impact prediction by in silico evolutionary lineage analysis strongly correlated with the measured phenotypic effect of each variant, providing a predictive tool for future studies on GPCR variants. Together, this large-scale functional study provides an operational framework for the postgenomic analysis of the multiple GPCR variants present in the human population. The association of T2D risk with signaling pathway-specific defects opens avenues for pathway-specific personalized therapeutic intervention and reveals the potential relevance of MT2 function during the day, when melatonin is undetectable, but spontaneous activity of the receptor occurs.
Collapse
Affiliation(s)
- Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Bianca Plouffe
- Institute for Research in Immunology and Cancer and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Québec H3C 3J7, Canada
| | - Amélie Bonnefond
- Université Lille, CNRS UMR 8199-EGID, Institut Pasteur de Lille, Lille, France
| | - Min Chen
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Jonathan Gallion
- Structural Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jean-Luc Guillaume
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Alan Hegron
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Mathilde Boissel
- Université Lille, CNRS UMR 8199-EGID, Institut Pasteur de Lille, Lille, France
| | - Mickaël Canouil
- Université Lille, CNRS UMR 8199-EGID, Institut Pasteur de Lille, Lille, France
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Québec H3C 3J7, Canada
| | - Viktoria Lukasheva
- Institute for Research in Immunology and Cancer and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Québec H3C 3J7, Canada
| | - Olivier Lichtarge
- Structural Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Philippe Froguel
- Université Lille, CNRS UMR 8199-EGID, Institut Pasteur de Lille, Lille, France. .,Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, W12 0NN London, UK
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Québec H3C 3J7, Canada.
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France. .,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
188
|
Fernandez TV, Leckman JF, Pittenger C. Genetic susceptibility in obsessive-compulsive disorder. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:767-781. [PMID: 29478613 DOI: 10.1016/b978-0-444-64076-5.00049-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Obsessive-compulsive disorder (OCD) is present in 1.5-2.5% of the population and can result in substantial lifelong disability. It is characterized by intrusive thoughts, sensations, and urges and by repetitive behaviors that are difficult to control despite, in most cases, preserved insight as to their excessive or irrational nature. The causes and underlying pathophysiology of OCD are not well understood, which has limited the development of new treatments and interventions. Despite evidence for a substantial genetic contribution to disease risk, identification and replication of genetic variants associated with OCD have been challenging. Decades of candidate gene association studies have provided little insight. They are now being supplanted by modern genomewide approaches to discover both common and rare sequence and structural variants. Studies to date suggest potential novel therapeutic avenues such as modulators of glutamatergic and immune pathways; however, individual genetic findings are not yet statistically robust or replicated. Further efforts are clearly needed to identify specific risk variants and to confirm vulnerable pathways by studying much larger cohorts of patients with comprehensive variant discovery approaches. Mouse knockout models have already made notable inroads into our understanding of OCD pathology; their utility will only increase as specific risk alleles are identified.
Collapse
Affiliation(s)
- Thomas V Fernandez
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.
| | - James F Leckman
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychology, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher Pittenger
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States; Department of Psychology, Yale University School of Medicine, New Haven, CT, United States; Integrated Neuroscience Research Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
189
|
He H, Holl K, DeBehnke S, Yeo CT, Hansen P, Gebre AK, Leone-Kabler S, Ruas M, Parks JS, Parrington J, Solberg Woods LC. Tpcn2 knockout mice have improved insulin sensitivity and are protected against high-fat diet-induced weight gain. Physiol Genomics 2018; 50:605-614. [PMID: 29750602 DOI: 10.1152/physiolgenomics.00135.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Type 2 diabetes is a complex disorder affected by multiple genes and the environment. Our laboratory has shown that in response to a glucose challenge, two-pore channel 2 ( Tpcn2) knockout mice exhibit a decreased insulin response but normal glucose clearance, suggesting they have improved insulin sensitivity compared with wild-type mice. We tested the hypothesis that improved insulin sensitivity in Tpcn2 knockout mice would protect against the negative effects of a high fat diet. Male and female Tpcn2 knockout (KO), heterozygous (Het), and wild-type (WT) mice were fed a low-fat (LF) or high-fat (HF) diet for 24 wk. HF diet significantly increases body weight in WT mice relative to those on the LF diet; this HF diet-induced increase in body weight is blunted in the Het and KO mice. Despite the protection against diet-induced weight gain, however, Tpcn2 KO mice are not protected against HF-diet-induced changes in glucose or insulin area under the curve during glucose tolerance tests in female mice, while HF diet has no significant effect on glucose tolerance in the male mice, regardless of genotype. Glucose disappearance during an insulin tolerance test is augmented in male KO mice, consistent with our previous findings suggesting enhanced insulin sensitivity in these mice. Male KO mice exhibit increased fasting plasma total cholesterol and triglyceride concentrations relative to WT mice on the LF diet, but this difference disappears in HF diet-fed mice where there is increased cholesterol and triglycerides across all genotypes. These data demonstrate that knockout of Tpcn2 may increase insulin action in male, but not female, mice. In addition, both male and female KO mice are protected against diet-induced weight gain, but this protection is likely independent from glucose tolerance, insulin sensitivity, and plasma lipid levels.
Collapse
Affiliation(s)
- Hong He
- Department of Pediatrics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Katie Holl
- Department of Pediatrics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Sarah DeBehnke
- Department of Pediatrics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Chay Teng Yeo
- Department of Pediatrics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Polly Hansen
- Department of Pediatrics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Abraham K Gebre
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina
| | - Sandra Leone-Kabler
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina
| | - Margarida Ruas
- Department of Pharmacology, University of Oxford , Oxford , United Kingdom
| | - John S Parks
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina
| | - John Parrington
- Department of Pharmacology, University of Oxford , Oxford , United Kingdom
| | - Leah C Solberg Woods
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina
| |
Collapse
|
190
|
Li Z, Chen P, Chen J, Xu Y, Wang Q, Li X, Li C, He L, Shi Y. Glucose and Insulin-Related Traits, Type 2 Diabetes and Risk of Schizophrenia: A Mendelian Randomization Study. EBioMedicine 2018; 34:182-188. [PMID: 30100396 PMCID: PMC6116472 DOI: 10.1016/j.ebiom.2018.07.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The link between schizophrenia and diabetes mellitus is well established by observational studies; however, the cause-effect relationship remains unclear. METHODS Here, we conducted Mendelian randomization analyses to assess a causal relationship of the genetic variants related to elevated fasting glucose levels, hemoglobin A1c (HbA1c), fasting insulin levels, and type 2 diabetes with the risk of schizophrenia. The analyses were performed using summary statistics obtained for the variants identified from the genome-wide association meta-analyses of fasting glucose levels (up to 133,010 individuals), HbA1c (up to 153,377 individuals), fasting insulin levels (up to 108,557 individuals), type 2 diabetes (up to 659,316 individuals), and schizophrenia (up to 108,341 individuals). The association between each variant and schizophrenia was weighted by its association with each studied condition, and estimates were combined using an inverse-variance weighted meta-analysis. FINDINGS Using information from thirteen variants related to fasting insulin levels, the causal effect of fasting insulin levels increases (per 1-SD) on the risk of schizophrenia was estimated at an odds ratio (OR) of 2·33 (p = 0·001), which is consistent with findings from the observational studies. The fasting glucose associated single nucleotide polymorphisms (SNPs) had no effect on the risk of schizophrenia in Europeans and East Asians (p > 0·05). Nonsignificant effects on the risk of schizophrenia was observed with raised HbA1c and type 2 diabetes, and consistent estimates were obtained across different populations. INTERPRETATION Our results suggest a causal role of elevated fasting insulin levels in schizophrenia pathogenesis.
Collapse
Affiliation(s)
- Zhiqiang Li
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China.
| | - Peng Chen
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, No. 45 Chaoyang Xi Road, Changchun 130021, PR China; College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun 130021, PR China; National Institute of Digestive, Diabetes and Kidney Diseases, National Institutes of Health, 445 N 5th St, Phoenix, AZ 85004, USA
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China
| | - Yifeng Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China
| | - Qingzhong Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Disease, The Metabolic Disease Institute of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China.
| |
Collapse
|
191
|
Onaolapo AY, Onaolapo OJ. Circadian dysrhythmia-linked diabetes mellitus: Examining melatonin’s roles in prophylaxis and management. World J Diabetes 2018; 9:99-114. [PMID: 30079146 PMCID: PMC6068738 DOI: 10.4239/wjd.v9.i7.99] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a chronic, life-threatening metabolic disorder that occurs worldwide. Despite an increase in the knowledge of the risk factors that are associated with diabetes mellitus, its worldwide prevalence has continued to rise; thus, necessitating more research into its aetiology. Recent researches are beginning to link a dysregulation of the circadian rhythm to impairment of intermediary metabolism; with evidences that circadian rhythm dysfunction might play an important role in the aetiology, course or prognosis of some cases of diabetes mellitus. These evidences thereby suggest possible relationships between the circadian rhythm regulator melatonin, and diabetes mellitus. In this review, we discuss the roles of the circadian rhythm in the regulation of the metabolism of carbohydrates and other macronutrients; with emphasis on the importance of melatonin and the impacts of its deficiency on carbohydrate homeostasis. Also, the possibility of using melatonin and its analogs for the “prophylaxis” or management of diabetes mellitus is also considered.
Collapse
Affiliation(s)
- Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho 210211, Oyo State, Nigeria
| | - Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo 230263, Osun State, Nigeria
| |
Collapse
|
192
|
Kanhaiya K, Rogojin V, Kazemi K, Czeizler E, Petre I. NetControl4BioMed: a pipeline for biomedical data acquisition and analysis of network controllability. BMC Bioinformatics 2018; 19:185. [PMID: 30066633 PMCID: PMC6069765 DOI: 10.1186/s12859-018-2177-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Network controllability focuses on discovering combinations of external interventions that can drive a biological system to a desired configuration. In practice, this approach translates into finding a combined multi-drug therapy in order to induce a desired response from a cell; this can lead to developments of novel therapeutic approaches for systemic diseases like cancer. RESULT We develop a novel bioinformatics data analysis pipeline called NetControl4BioMed based on the concept of target structural control of linear networks. Our pipeline generates novel molecular interaction networks by combining pathway data from various public databases starting from the user's query. The pipeline then identifies a set of nodes that is enough to control a given, user-defined set of disease-specific essential proteins in the network, i.e., it is able to induce a change in their configuration from any initial state to any final state. We provide both the source code of the pipeline as well as an online web-service based on this pipeline http://combio.abo.fi/nc/net_control/remote_call.php . CONCLUSION The pipeline can be used by researchers for controlling and better understanding of molecular interaction networks through combinatorial multi-drug therapies, for more efficient therapeutic approaches and personalised medicine.
Collapse
Affiliation(s)
- Krishna Kanhaiya
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Vladimir Rogojin
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Keivan Kazemi
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Eugen Czeizler
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
- National Institute for Research and Development for Biological Sciences, Splaiul Independentei 296, Bucharest, 060031 Romania
| | - Ion Petre
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
- National Institute for Research and Development for Biological Sciences, Splaiul Independentei 296, Bucharest, 060031 Romania
| |
Collapse
|
193
|
Perez-Alcantara M, Honoré C, Wesolowska-Andersen A, Gloyn AL, McCarthy MI, Hansson M, Beer NL, van de Bunt M. Patterns of differential gene expression in a cellular model of human islet development, and relationship to type 2 diabetes predisposition. Diabetologia 2018; 61:1614-1622. [PMID: 29675560 PMCID: PMC6354904 DOI: 10.1007/s00125-018-4612-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/12/2018] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Most type 2 diabetes-associated genetic variants identified via genome-wide association studies (GWASs) appear to act via the pancreatic islet. Observed defects in insulin secretion could result from an impact of these variants on islet development and/or the function of mature islets. Most functional studies have focused on the latter, given limitations regarding access to human fetal islet tissue. Capitalising upon advances in in vitro differentiation, we characterised the transcriptomes of human induced pluripotent stem cell (iPSC) lines differentiated along the pancreatic endocrine lineage, and explored the contribution of altered islet development to the pathogenesis of type 2 diabetes. METHODS We performed whole-transcriptome RNA sequencing of human iPSC lines from three independent donors, at baseline and at seven subsequent stages during in vitro islet differentiation. Differentially expressed genes (q < 0.01, log2 fold change [FC] > 1) were assigned to the stages at which they were most markedly upregulated. We used these data to characterise upstream transcription factors directing different stages of development, and to explore the relationship between RNA expression profiles and genes mapping to type 2 diabetes GWAS signals. RESULTS We identified 9409 differentially expressed genes across all stages, including many known markers of islet development. Integration of differential expression data with information on transcription factor motifs highlighted the potential contribution of REST to islet development. Over 70% of genes mapping within type 2 diabetes-associated credible intervals showed peak differential expression during islet development, and type 2 diabetes GWAS loci of largest effect (including TCF7L2; log2FC = 1.2; q = 8.5 × 10-10) were notably enriched in genes differentially expressed at the posterior foregut stage (q = 0.002), as calculated by gene set enrichment analyses. In a complementary analysis of enrichment, genes differentially expressed in the final, beta-like cell stage of in vitro differentiation were significantly enriched (hypergeometric test, permuted p value <0.05) for genes within the credible intervals of type 2 diabetes GWAS loci. CONCLUSIONS/INTERPRETATION The present study characterises RNA expression profiles during human islet differentiation, identifies potential transcriptional regulators of the differentiation process, and suggests that the inherited predisposition to type 2 diabetes is partly mediated through modulation of islet development. DATA AVAILABILITY Sequence data for this study has been deposited at the European Genome-phenome Archive (EGA), under accession number EGAS00001002721.
Collapse
Affiliation(s)
| | - Christian Honoré
- Department of Stem Cell Biology, Novo Nordisk A/S, Maaloev, Denmark
| | | | - Anna L Gloyn
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Old Road, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Old Road, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Mattias Hansson
- Department of Stem Cell Research, Novo Nordisk A/S, Maaloev, Denmark
| | - Nicola L Beer
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Old Road, Oxford, OX3 7LE, UK.
| | - Martijn van de Bunt
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Old Road, Oxford, OX3 7LE, UK
| |
Collapse
|
194
|
Nind T, Galloway J, McAllister G, Scobbie D, Bonney W, Hall C, Tramma L, Reel P, Groves M, Appleby P, Doney A, Guthrie B, Jefferson E. The research data management platform (RDMP): A novel, process driven, open-source tool for the management of longitudinal cohorts of clinical data. Gigascience 2018; 7:5001426. [PMID: 29790950 PMCID: PMC6041881 DOI: 10.1093/gigascience/giy060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 11/13/2022] Open
Abstract
Background The Health Informatics Centre at the University of Dundee provides a service to securely host clinical datasets and extract relevant data for anonymized cohorts to researchers to enable them to answer key research questions. As is common in research using routine healthcare data, the service was historically delivered using ad-hoc processes resulting in the slow provision of data whose provenance was often hidden to the researchers using it. This paper describes the development and evaluation of the Research Data Management Platform (RDMP): an open source tool to load, manage, clean, and curate longitudinal healthcare data for research and provide reproducible and updateable datasets for defined cohorts to researchers. Results Between 2013 and 2017, RDMP tool implementation tripled the productivity of data analysts producing data releases for researchers from 7.1 to 25.3 per month and reduced the error rate from 12.7% to 3.1%. The effort on data management reduced from a mean of 24.6 to 3.0 hours per data release. The waiting time for researchers to receive data after agreeing a specification reduced from approximately 6 months to less than 1 week. The software is scalable and currently manages 163 datasets. A total 1,321 data extracts for research have been produced, with the largest extract linking data from 70 different datasets. Conclusions The tools and processes that encompass the RDMP not only fulfil the research data management requirements of researchers but also support the seamless collaboration of data cleaning, data transformation, data summarization and data quality assessment activities by different research groups.
Collapse
Affiliation(s)
- Thomas Nind
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - James Galloway
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Gordon McAllister
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Donald Scobbie
- Edinburgh Parallel Computing Centre, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh, EH9 3FD, UK
| | - Wilfred Bonney
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Christopher Hall
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Leandro Tramma
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Parminder Reel
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Martin Groves
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Philip Appleby
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Alex Doney
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Bruce Guthrie
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Emily Jefferson
- Health Informatics Centre, University of Dundee, Mail Box 15, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| |
Collapse
|
195
|
Strawbridge RJ, van Zuydam NR. Shared Genetic Contribution of Type 2 Diabetes and Cardiovascular Disease: Implications for Prognosis and Treatment. Curr Diab Rep 2018; 18:59. [PMID: 29938349 PMCID: PMC6015804 DOI: 10.1007/s11892-018-1021-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW The increased cardiovascular disease (CVD) risk in subjects with type 2 diabetes (T2D) is well established. This review collates the available evidence and assesses the shared genetic background between T2D and CVD: the causal contribution of common risk factors to T2D and CVD and how genetics can be used to improve drug development and clinical outcomes. RECENT FINDINGS Large-scale genome-wide association studies (GWAS) of T2D and CVD support a shared genetic background but minimal individual locus overlap. Mendelian randomisation (MR) analyses show that T2D is causal for CVD, but GWAS of CVD, T2D and their common risk factors provided limited evidence for individual locus overlap. Distinct but functionally related pathways were enriched for CVD and T2D genetic associations reflecting the lack of locus overlap and providing some explanation for the variable associations of common risk factors with CVD and T2D from MR analyses.
Collapse
Affiliation(s)
- Rona J. Strawbridge
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Room 113, 1 Lilybank Gardens, Glasgow, G12 8RZ UK
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Natalie R. van Zuydam
- Wellcome Centre Human Genetics, University of Oxford, Roosevelt Drive, Headington, Oxford, Oxfordshire, OX3 7BN UK
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Headington, Oxford, Oxfordshire, OX3 7LE UK
| |
Collapse
|
196
|
Langenberg C, Lotta LA. Genomic insights into the causes of type 2 diabetes. Lancet 2018; 391:2463-2474. [PMID: 29916387 DOI: 10.1016/s0140-6736(18)31132-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/30/2018] [Accepted: 05/15/2018] [Indexed: 01/05/2023]
Abstract
Genome-wide association studies have implicated around 250 genomic regions in predisposition to type 2 diabetes, with evidence for causal variants and genes emerging for several of these regions. Understanding of the underlying mechanisms, including the interplay between β-cell failure, insulin sensitivity, appetite regulation, and adipose storage has been facilitated by the integration of multidimensional data for diabetes-related intermediate phenotypes, detailed genomic annotations, functional experiments, and now multiomic molecular features. Studies in diverse ethnic groups and examples from population isolates have shown the value and need for a broad genomic approach to this global disease. Transethnic discovery efforts and large-scale biobanks in diverse populations and ancestries could help to address some of the Eurocentric bias. Despite rapid progress in the discovery of the highly polygenic architecture of type 2 diabetes, dominated by common alleles with small, cumulative effects on disease risk, these insights have been of little clinical use in terms of disease prediction or prevention, and have made only small contributions to subtype classification or stratified approaches to treatment. Successful development of academia-industry partnerships for exome or genome sequencing in large biobanks could help to deliver economies of scale, with implications for the future of genomics-focused research.
Collapse
Affiliation(s)
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
197
|
Morris AP. Progress in defining the genetic contribution to type 2 diabetes susceptibility. Curr Opin Genet Dev 2018; 50:41-51. [DOI: 10.1016/j.gde.2018.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/30/2022]
|
198
|
Abstract
Endocrine organs secrete a variety of hormones involved in the regulation of a multitude of body functions. Although pancreatic islets were discovered at the turn of the 19th century, other endocrine glands remained commonly described as diffuse endocrine systems. Over the last two decades, development of new imaging techniques and genetically-modified animals with cell-specific fluorescent tags or specific hormone deficiencies have enabled in vivo imaging of endocrine organs and revealed intricate endocrine cell network structures and plasticity. Overall, these new tools have revolutionized our understanding of endocrine function. The overarching aim of this Review is to describe the current mechanistic understanding that has emerged from imaging studies of endocrine cell network structure/function relationships in animal models, with a particular emphasis on the pituitary gland and the endocrine pancreas.
Collapse
Affiliation(s)
- Patrice Mollard
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, F-34094, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, F-34094, Montpellier, France.
| |
Collapse
|
199
|
Fawad A, Bergmann A, Struck J, Nilsson PM, Orho-Melander M, Melander O. Proneurotensin Predicts Cardiovascular Disease in an Elderly Population. J Clin Endocrinol Metab 2018; 103:1940-1947. [PMID: 29546332 DOI: 10.1210/jc.2017-02424] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/07/2018] [Indexed: 01/28/2023]
Abstract
CONTEXT The gut hormone neurotensin promotes fat absorption, diet-induced weight gain, and liver steatosis. Its stable precursor-hormone fragment "proneurotensin" predicts cardiometabolic disease in middle-aged populations, especially in women. OBJECTIVE To test if proneurotensin predicts cardiovascular disease (CVD) and diabetes development in an elderly population and whether there are gender differences in this respect. DESIGN, SETTING, AND PARTICIPANTS Fasting proneurotensin was measured in plasma from 4804 participants (mean age 69 ± 6 years) of the Malmö Preventive Project and subjects were followed up for development of CVD and diabetes during 5.4 years. MAIN OUTCOME MEASURES Multivariate adjusted Cox proportional hazard models CVD were used to relate the proneurotensin to the risk of incident CVD and diabetes in all subjects and in gender-stratified analyses. RESULTS In total, there were 456 first CVD events and 222 incident cases of diabetes. The hazard ratio [HR (95% confidence interval)] for CVD per 1 standard deviation (SD) increment of proneurotensin was 1.10 (1.01 to 1.21); P = 0.037, and the above vs below median HR was 1.27 (1.06 to 1.54); P = 0.011, with similar effect sizes in both genders. There was no significant association between proneurotensin and incident diabetes in the entire population (P = 0.52) or among men (P = 0.52). However, in women proneurotensin predicted diabetes incidence with a per 1 SD increment HR of 1.28 (1.30 to 1.59); P = 0.025 and an above vs below median HR of 1.41 (1.10 to 1.80); P = 0.007. CONCLUSIONS In the elderly population, proneurotensin independently predicts development of CVD in both genders, whereas it only predicts diabetes in women.
Collapse
Affiliation(s)
- Ayesha Fawad
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Andreas Bergmann
- Sphingotec GmbH, Hennigsdorf, Germany
- Waltraut Bergmann Foundation, Hohen Neuendorf, Germany
| | - Joachim Struck
- Sphingotec GmbH, Hennigsdorf, Germany
- Waltraut Bergmann Foundation, Hohen Neuendorf, Germany
| | - Peter M Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
200
|
The molecular functions of hepatocyte nuclear factors - In and beyond the liver. J Hepatol 2018; 68:1033-1048. [PMID: 29175243 DOI: 10.1016/j.jhep.2017.11.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/27/2022]
Abstract
The hepatocyte nuclear factors (HNFs) namely HNF1α/β, FOXA1/2/3, HNF4α/γ and ONECUT1/2 are expressed in a variety of tissues and organs, including the liver, pancreas and kidney. The spatial and temporal manner of HNF expression regulates embryonic development and subsequently the development of multiple tissues during adulthood. Though the HNFs were initially identified individually based on their roles in the liver, numerous studies have now revealed that the HNFs cross-regulate one another and exhibit synergistic relationships in the regulation of tissue development and function. The complex HNF transcriptional regulatory networks have largely been elucidated in rodent models, but less so in human biological systems. Several heterozygous mutations in these HNFs were found to cause diseases in humans but not in rodents, suggesting clear species-specific differences in mutational mechanisms that remain to be uncovered. In this review, we compare and contrast the expression patterns of the HNFs, the HNF cross-regulatory networks and how these liver-enriched transcription factors serve multiple functions in the liver and beyond, extending our focus to the pancreas and kidney. We also summarise the insights gained from both human and rodent studies of mutations in several HNFs that are known to lead to different disease conditions.
Collapse
|