251
|
Navarro G, Abdolazimi Y, Zhao Z, Xu H, Lee S, Armstrong NA, Annes JP. Genetic Disruption of Adenosine Kinase in Mouse Pancreatic β-Cells Protects Against High-Fat Diet-Induced Glucose Intolerance. Diabetes 2017; 66:1928-1938. [PMID: 28468960 PMCID: PMC5482077 DOI: 10.2337/db16-0816] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 04/24/2017] [Indexed: 01/03/2023]
Abstract
Islet β-cells adapt to insulin resistance through increased insulin secretion and expansion. Type 2 diabetes typically occurs when prolonged insulin resistance exceeds the adaptive capacity of β-cells. Our prior screening efforts led to the discovery that adenosine kinase (ADK) inhibitors stimulate β-cell replication. Here, we evaluated whether ADK disruption in mouse β-cells affects β-cell mass and/or protects against high-fat diet (HFD)-induced glucose dysregulation. Mice targeted at the Adk locus were bred to Rip-Cre and Ins1-Cre/ERT1Lphi mice to enable constitutive (βADKO) and conditional (iβADKO) disruption of ADK expression in β-cells, respectively. Weight gain, glucose tolerance, insulin sensitivity, and glucose-stimulated insulin secretion (GSIS) were longitudinally monitored in normal chow (NC)-fed and HFD-fed mice. In addition, β-cell mass and replication were measured by immunofluorescence-based islet morphometry. NC-fed adult βADKO and iβADKO mice displayed glucose tolerance, insulin tolerance and β-cell mass comparable to control animals. By contrast, HFD-fed βADKO and iβADKO animals had improved glucose tolerance and increased in vivo GSIS. Improved glucose handling was associated with increased β-cell replication and mass. We conclude that ADK expression negatively regulates the adaptive β-cell response to HFD challenge. Therefore, modulation of ADK activity is a potential strategy for enhancing the adaptive β-cell response.
Collapse
Affiliation(s)
- Guadalupe Navarro
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| | - Yassan Abdolazimi
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| | - Zhengshan Zhao
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| | - Haixia Xu
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Sooyeon Lee
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| | - Neali A Armstrong
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| | - Justin P Annes
- Department of Medicine and Division of Endocrinology, Stanford University, Stanford, CA
| |
Collapse
|
252
|
Nikolic M, Papantonis A, Rada-Iglesias A. GARLIC: a bioinformatic toolkit for aetiologically connecting diseases and cell type-specific regulatory maps. Hum Mol Genet 2017; 26:742-752. [PMID: 28007912 PMCID: PMC5409087 DOI: 10.1093/hmg/ddw423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 12/12/2016] [Indexed: 12/28/2022] Open
Abstract
Genome-wide association studies (GWAS) have emerged as a powerful tool to uncover the genetic basis of human common diseases, which often show a complex, polygenic and multi-factorial aetiology. These studies have revealed that 70–90% of all single nucleotide polymorphisms (SNPs) associated with common complex diseases do not occur within genes (i.e. they are non-coding), making the discovery of disease-causative genetic variants and the elucidation of the underlying pathological mechanisms far from straightforward. Based on emerging evidences suggesting that disease-associated SNPs are frequently found within cell type-specific regulatory sequences, here we present GARLIC (GWAS-based Prediction Toolkit for Connecting Diseases and Cell Types), a user-friendly, multi-purpose software with an associated database and online viewer that, using global maps of cis-regulatory elements, can aetiologically connect human diseases with relevant cell types. Additionally, GARLIC can be used to retrieve potential disease-causative genetic variants overlapping regulatory sequences of interest. Overall, GARLIC can satisfy several important needs within the field of medical genetics, thus potentially assisting in the ultimate goal of uncovering the elusive and complex genetic basis of common human disorders.
Collapse
Affiliation(s)
- Miloš Nikolic
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931 Cologne, Germany.,The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Argyris Papantonis
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Alvaro Rada-Iglesias
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931 Cologne, Germany.,The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| |
Collapse
|
253
|
Thomsen SK, Gloyn AL. Human genetics as a model for target validation: finding new therapies for diabetes. Diabetologia 2017; 60:960-970. [PMID: 28447115 PMCID: PMC5423999 DOI: 10.1007/s00125-017-4270-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/14/2017] [Indexed: 01/01/2023]
Abstract
Type 2 diabetes is a global epidemic with major effects on healthcare expenditure and quality of life. Currently available treatments are inadequate for the prevention of comorbidities, yet progress towards new therapies remains slow. A major barrier is the insufficiency of traditional preclinical models for predicting drug efficacy and safety. Human genetics offers a complementary model to assess causal mechanisms for target validation. Genetic perturbations are 'experiments of nature' that provide a uniquely relevant window into the long-term effects of modulating specific targets. Here, we show that genetic discoveries over the past decades have accurately predicted (now known) therapeutic mechanisms for type 2 diabetes. These findings highlight the potential for use of human genetic variation for prospective target validation, and establish a framework for future applications. Studies into rare, monogenic forms of diabetes have also provided proof-of-principle for precision medicine, and the applicability of this paradigm to complex disease is discussed. Finally, we highlight some of the limitations that are relevant to the use of genome-wide association studies (GWAS) in the search for new therapies for diabetes. A key outstanding challenge is the translation of GWAS signals into disease biology and we outline possible solutions for tackling this experimental bottleneck.
Collapse
Affiliation(s)
- Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
- National Institute of Health Research Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
254
|
Aung T, Ozaki M, Lee MC, Schlötzer-Schrehardt U, Thorleifsson G, Mizoguchi T, Igo RP, Haripriya A, Williams SE, Astakhov YS, Orr AC, Burdon KP, Nakano S, Mori K, Abu-Amero K, Hauser M, Li Z, Prakadeeswari G, Bailey JNC, Cherecheanu AP, Kang JH, Nelson S, Hayashi K, Manabe SI, Kazama S, Zarnowski T, Inoue K, Irkec M, Coca-Prados M, Sugiyama K, Järvelä I, Schlottmann P, Lerner SF, Lamari H, Nilgün Y, Bikbov M, Park KH, Cha SC, Yamashiro K, Zenteno JC, Jonas JB, Kumar RS, Perera SA, Chan ASY, Kobakhidze N, George R, Vijaya L, Do T, Edward DP, de Juan Marcos L, Pakravan M, Moghimi S, Ideta R, Bach-Holm D, Kappelgaard P, Wirostko B, Thomas S, Gaston D, Bedard K, Greer WL, Yang Z, Chen X, Huang L, Sang J, Jia H, Jia L, Qiao C, Zhang H, Liu X, Zhao B, Wang YX, Xu L, Leruez S, Reynier P, Chichua G, Tabagari S, Uebe S, Zenkel M, Berner D, Mossböck G, Weisschuh N, Hoja U, Welge-Luessen UC, Mardin C, Founti P, Chatzikyriakidou A, Pappas T, Anastasopoulos E, Lambropoulos A, Ghosh A, Shetty R, Porporato N, Saravanan V, Venkatesh R, Shivkumar C, Kalpana N, Sarangapani S, Kanavi MR, Beni AN, Yazdani S, et alAung T, Ozaki M, Lee MC, Schlötzer-Schrehardt U, Thorleifsson G, Mizoguchi T, Igo RP, Haripriya A, Williams SE, Astakhov YS, Orr AC, Burdon KP, Nakano S, Mori K, Abu-Amero K, Hauser M, Li Z, Prakadeeswari G, Bailey JNC, Cherecheanu AP, Kang JH, Nelson S, Hayashi K, Manabe SI, Kazama S, Zarnowski T, Inoue K, Irkec M, Coca-Prados M, Sugiyama K, Järvelä I, Schlottmann P, Lerner SF, Lamari H, Nilgün Y, Bikbov M, Park KH, Cha SC, Yamashiro K, Zenteno JC, Jonas JB, Kumar RS, Perera SA, Chan ASY, Kobakhidze N, George R, Vijaya L, Do T, Edward DP, de Juan Marcos L, Pakravan M, Moghimi S, Ideta R, Bach-Holm D, Kappelgaard P, Wirostko B, Thomas S, Gaston D, Bedard K, Greer WL, Yang Z, Chen X, Huang L, Sang J, Jia H, Jia L, Qiao C, Zhang H, Liu X, Zhao B, Wang YX, Xu L, Leruez S, Reynier P, Chichua G, Tabagari S, Uebe S, Zenkel M, Berner D, Mossböck G, Weisschuh N, Hoja U, Welge-Luessen UC, Mardin C, Founti P, Chatzikyriakidou A, Pappas T, Anastasopoulos E, Lambropoulos A, Ghosh A, Shetty R, Porporato N, Saravanan V, Venkatesh R, Shivkumar C, Kalpana N, Sarangapani S, Kanavi MR, Beni AN, Yazdani S, Lashay A, Naderifar H, Khatibi N, Fea A, Lavia C, Dallorto L, Rolle T, Frezzotti P, Paoli D, Salvi E, Manunta P, Mori Y, Miyata K, Higashide T, Chihara E, Ishiko S, Yoshida A, Yanagi M, Kiuchi Y, Ohashi T, Sakurai T, Sugimoto T, Chuman H, Aihara M, Inatani M, Miyake M, Gotoh N, Matsuda F, Yoshimura N, Ikeda Y, Ueno M, Sotozono C, Jeoung JW, Sagong M, Park KH, Ahn J, Cruz-Aguilar M, Ezzouhairi SM, Rafei A, Chong YF, Ng XY, Goh SR, Chen Y, Yong VHK, Khan MI, Olawoye OO, Ashaye AO, Ugbede I, Onakoya A, Kizor-Akaraiwe N, Teekhasaenee C, Suwan Y, Supakontanasan W, Okeke S, Uche NJ, Asimadu I, Ayub H, Akhtar F, Kosior-Jarecka E, Lukasik U, Lischinsky I, Castro V, Grossmann RP, Sunaric Megevand G, Roy S, Dervan E, Silke E, Rao A, Sahay P, Fornero P, Cuello O, Sivori D, Zompa T, Mills RA, Souzeau E, Mitchell P, Wang JJ, Hewitt AW, Coote M, Crowston JG, Astakhov SY, Akopov EL, Emelyanov A, Vysochinskaya V, Kazakbaeva G, Fayzrakhmanov R, Al-Obeidan SA, Owaidhah O, Aljasim LA, Chowbay B, Foo JN, Soh RQ, Sim KS, Xie Z, Cheong AWO, Mok SQ, Soo HM, Chen XY, Peh SQ, Heng KK, Husain R, Ho SL, Hillmer AM, Cheng CY, Escudero-Domínguez FA, González-Sarmiento R, Martinon-Torres F, Salas A, Pathanapitoon K, Hansapinyo L, Wanichwecharugruang B, Kitnarong N, Sakuntabhai A, Nguyn HX, Nguyn GTT, Nguyn TV, Zenz W, Binder A, Klobassa DS, Hibberd ML, Davila S, Herms S, Nöthen MM, Moebus S, Rautenbach RM, Ziskind A, Carmichael TR, Ramsay M, Álvarez L, García M, González-Iglesias H, Rodríguez-Calvo PP, Fernández-Vega Cueto L, Oguz Ç, Tamcelik N, Atalay E, Batu B, Aktas D, Kasım B, Wilson MR, Coleman AL, Liu Y, Challa P, Herndon L, Kuchtey RW, Kuchtey J, Curtin K, Chaya CJ, Crandall A, Zangwill LM, Wong TY, Nakano M, Kinoshita S, den Hollander AI, Vesti E, Fingert JH, Lee RK, Sit AJ, Shingleton BJ, Wang N, Cusi D, Qamar R, Kraft P, Pericak-Vance MA, Raychaudhuri S, Heegaard S, Kivelä T, Reis A, Kruse FE, Weinreb RN, Pasquale LR, Haines JL, Thorsteinsdottir U, Jonasson F, Allingham RR, Milea D, Ritch R, Kubota T, Tashiro K, Vithana EN, Micheal S, Topouzis F, Craig JE, Dubina M, Sundaresan P, Stefansson K, Wiggs JL, Pasutto F, Khor CC. Genetic association study of exfoliation syndrome identifies a protective rare variant at LOXL1 and five new susceptibility loci. Nat Genet 2017; 49:993-1004. [PMID: 28553957 DOI: 10.1038/ng.3875] [Show More Authors] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/26/2017] [Indexed: 12/14/2022]
Abstract
Exfoliation syndrome (XFS) is the most common known risk factor for secondary glaucoma and a major cause of blindness worldwide. Variants in two genes, LOXL1 and CACNA1A, have previously been associated with XFS. To further elucidate the genetic basis of XFS, we collected a global sample of XFS cases to refine the association at LOXL1, which previously showed inconsistent results across populations, and to identify new variants associated with XFS. We identified a rare protective allele at LOXL1 (p.Phe407, odds ratio (OR) = 25, P = 2.9 × 10-14) through deep resequencing of XFS cases and controls from nine countries. A genome-wide association study (GWAS) of XFS cases and controls from 24 countries followed by replication in 18 countries identified seven genome-wide significant loci (P < 5 × 10-8). We identified association signals at 13q12 (POMP), 11q23.3 (TMEM136), 6p21 (AGPAT1), 3p24 (RBMS3) and 5q23 (near SEMA6A). These findings provide biological insights into the pathology of XFS and highlight a potential role for naturally occurring rare LOXL1 variants in disease biology.
Collapse
Affiliation(s)
- Tin Aung
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mineo Ozaki
- Ozaki Eye Hospital, Hyuga, Miyazaki, Japan.,Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Mei Chin Lee
- Singapore Eye Research Institute, Singapore.,Academic Clinical Program for Ophthalmology and Visual Sciences, Office of Clinical and Academic Faculty Affairs, Duke-NUS Graduate Medical School, Singapore
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Robert P Igo
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Susan E Williams
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Yury S Astakhov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Andrew C Orr
- Department of Ophthalmology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathryn P Burdon
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Satoko Nakano
- Department of Ophthalmology, Oita University Faculty of Medicine, Oita, Japan
| | - Kazuhiko Mori
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Khaled Abu-Amero
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Department of Ophthalmology, College of Medicine, University of Florida, Jacksonville, Florida, USA
| | - Michael Hauser
- Singapore Eye Research Institute, Singapore.,Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Zheng Li
- Genome Institute of Singapore, Singapore
| | | | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Alina Popa Cherecheanu
- 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.,Department of Ophthalmology, University Emergency Hospital, Bucharest, Romania
| | - Jae H Kang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Nelson
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | | | | | | | - Tomasz Zarnowski
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University, Lublin, Poland
| | | | - Murat Irkec
- Department of Ophthalmology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Miguel Coca-Prados
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain.,Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazuhisa Sugiyama
- Department of Ophthalmology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Irma Järvelä
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | | | - S Fabian Lerner
- Fundación para el Estudio del Glaucoma, Buenos Aires, Argentina
| | - Hasnaa Lamari
- Clinique Spécialisée en Ophtalmologie Mohammedia, Mohammedia, Morocco
| | - Yildirim Nilgün
- Department of Ophthalmology, Eskisehir Osmangazi University, Meselik, Eskisehir, Turkey
| | | | - Ki Ho Park
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soon Cheol Cha
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Ophthalmology, Otsu Red Cross Hospital, Otsu, Japan
| | - Juan C Zenteno
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico.,Biochemistry Department, Faculty of Medicine, UNAM, Mexico City, Mexico
| | - Jost B Jonas
- Department of Ophthalmology, Medical Faculty Mannheim of the Ruprecht Karls University of Heidelberg, Mannheim, Germany.,Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | | | - Shamira A Perera
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore
| | - Anita S Y Chan
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Academic Clinical Program for Ophthalmology and Visual Sciences, Office of Clinical and Academic Faculty Affairs, Duke-NUS Graduate Medical School, Singapore
| | | | - Ronnie George
- Jadhavbhai Nathamal Singhvi Department of Glaucoma, Medical Research Foundation, Chennai, India
| | - Lingam Vijaya
- Jadhavbhai Nathamal Singhvi Department of Glaucoma, Medical Research Foundation, Chennai, India
| | - Tan Do
- Vietnam National Institute of Ophthalmology, Hanoi, Vietnam
| | - Deepak P Edward
- King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia.,Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lourdes de Juan Marcos
- Department of Ophthalmology, University Hospital of Salamanca, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Mohammad Pakravan
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sasan Moghimi
- Farabi Eye Hospital, Tehran University Eye Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Barbara Wirostko
- John A. Moran Eye Center, Department of Ophthalmology, University of Utah, Salt Lake City, Utah, USA
| | - Samuel Thomas
- John A. Moran Eye Center, Department of Ophthalmology, University of Utah, Salt Lake City, Utah, USA
| | - Daniel Gaston
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Karen Bedard
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Wenda L Greer
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xueyi Chen
- Department of Ophthalmology, First Affiliated Hospital of Xinjiang Medical University, Urumchi, China
| | - Lulin Huang
- Center for Human Molecular Biology and Genetics, Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China.,Sichuan Translational Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Jinghong Sang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Hongyan Jia
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Liyun Jia
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China.,Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chunyan Qiao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Hui Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Xuyang Liu
- Shenzhen Key Laboratory of Ophthalmology, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| | - Bowen Zhao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China.,Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ya-Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liang Xu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Stéphanie Leruez
- Département d'Ophtalmologie, Centre Hospitalier Universitaire, Angers, France
| | - Pascal Reynier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
| | | | | | - Steffen Uebe
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel Berner
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Georg Mossböck
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - Nicole Weisschuh
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Ursula Hoja
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrich-Christoph Welge-Luessen
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Mardin
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Panayiota Founti
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anthi Chatzikyriakidou
- Laboratory of General Biology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theofanis Pappas
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios Anastasopoulos
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Lambropoulos
- Laboratory of General Biology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Rohit Shetty
- Narayana Nethralaya Eye Hospital, Bangalore, India
| | | | - Vijayan Saravanan
- Department of Genetics, Aravind Medical Research Foundation, Madurai, India
| | | | | | | | | | - Mozhgan R Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Naderi Beni
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Yazdani
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Lashay
- Farabi Eye Hospital, Tehran University Eye Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Naderifar
- Farabi Eye Hospital, Tehran University Eye Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nassim Khatibi
- Farabi Eye Hospital, Tehran University Eye Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Antonio Fea
- Dipartimento di Scienze Chirurgiche, Università di Torino, Turin, Italy
| | - Carlo Lavia
- Dipartimento di Scienze Chirurgiche, Università di Torino, Turin, Italy
| | - Laura Dallorto
- Dipartimento di Scienze Chirurgiche, Università di Torino, Turin, Italy
| | - Teresa Rolle
- Dipartimento di Scienze Chirurgiche, Università di Torino, Turin, Italy
| | - Paolo Frezzotti
- Ophthalmology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Daniela Paoli
- Department of Ophthalmology, Monfalcone Hospital, Gorizia, Italy
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Paolo Manunta
- Department of Nephrology, University Vita-Salute San Raffaele, Milan, Italy
| | | | | | - Tomomi Higashide
- Department of Ophthalmology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | | | - Satoshi Ishiko
- Department of Medicine and Engineering Combined Research Institute, Asahikawa Medical University, Asahikawa, Japan
| | - Akitoshi Yoshida
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Masahide Yanagi
- Department of Ophthalmology and Visual Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology and Visual Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | - Takako Sugimoto
- Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideki Chuman
- Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Makoto Aihara
- Department of Ophthalmology, University of Tokyo, Tokyo, Japan
| | - Masaru Inatani
- Department of Ophthalmology, Faculty of Medical Science, University of Fukui, Fukui, Japan
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norimoto Gotoh
- Center for Genomic Medicine, INSERM U852, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, INSERM U852, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nagahisa Yoshimura
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Tazuke Kofukai Foundation, Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Yoko Ikeda
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Morio Ueno
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jin Wook Jeoung
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Sagong
- Department of Ophthalmology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University Bundang Hospital, Gyeonggi, Republic of Korea
| | - Jeeyun Ahn
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Marisa Cruz-Aguilar
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico
| | - Sidi M Ezzouhairi
- Clinique Spécialisée en Ophtalmologie Mohammedia, Mohammedia, Morocco
| | | | | | - Xiao Yu Ng
- Singapore Eye Research Institute, Singapore
| | | | | | | | - Muhammad Imran Khan
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Olusola O Olawoye
- Department of Ophthalmology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Department of Ophthalmology, University College Hospital, Ibadan, Nigeria
| | - Adeyinka O Ashaye
- Department of Ophthalmology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Department of Ophthalmology, University College Hospital, Ibadan, Nigeria
| | | | - Adeola Onakoya
- Department of Ophthalmology, University of Lagos, Lagos, Nigeria.,Guinness Eye Centre, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Nkiru Kizor-Akaraiwe
- Department of Ophthalmology, ESUT Teaching Hospital Parklane, Enugu, Nigeria.,Eye Specialists Hospital, Enugu, Nigeria
| | - Chaiwat Teekhasaenee
- Department of Ophthalmology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Yanin Suwan
- Department of Ophthalmology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wasu Supakontanasan
- Department of Ophthalmology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suhanya Okeke
- Department of Ophthalmology, ESUT Teaching Hospital Parklane, Enugu, Nigeria.,Eye Specialists Hospital, Enugu, Nigeria
| | - Nkechi J Uche
- Eye Specialists Hospital, Enugu, Nigeria.,Department of Ophthalmology, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria.,Department of Ophthalmology, College of Medicine, University of Nigeria, Nsukka, Ituku Ozalla Campus, Enugu, Nigeria
| | - Ifeoma Asimadu
- Department of Ophthalmology, ESUT Teaching Hospital Parklane, Enugu, Nigeria
| | - Humaira Ayub
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Abbottabad, Pakistan
| | - Farah Akhtar
- Pakistan Institute of Ophthalmology, Al-Shifa Trust Eye Hospital, Rawalpindi, Pakistan
| | - Ewa Kosior-Jarecka
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University, Lublin, Poland
| | - Urszula Lukasik
- Department of Diagnostics and Microsurgery of Glaucoma, Medical University, Lublin, Poland
| | | | - Vania Castro
- Universidad Peruana Cayetano Heredia, Hospital Nacional Arzobispo Loayza, Lima, Peru
| | | | - Gordana Sunaric Megevand
- Clinical Research Centre Adolphe de Rothschild, Société Médicale de Beaulieu, Geneva, Switzerland
| | - Sylvain Roy
- Clinical Research Centre Adolphe de Rothschild, Société Médicale de Beaulieu, Geneva, Switzerland
| | - Edward Dervan
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eoin Silke
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Aparna Rao
- Shri Mithu Tulsi, LV Prasad Eye Institute, Bhubaneswar, India
| | - Priti Sahay
- Shri Mithu Tulsi, LV Prasad Eye Institute, Bhubaneswar, India
| | | | | | - Delia Sivori
- Fundación para el Estudio del Glaucoma, Buenos Aires, Argentina
| | - Tamara Zompa
- Centro Oftalmologico Charles, Buenos Aires, Argentina
| | - Richard A Mills
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Emmanuelle Souzeau
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Paul Mitchell
- Centre for Vision Research, Department of Ophthalmology and Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Jie Jin Wang
- Centre for Vision Research, Department of Ophthalmology and Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Alex W Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Centre for Eye Research Australia (CERA), University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Michael Coote
- Centre for Eye Research Australia (CERA), University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Jonathan G Crowston
- Centre for Eye Research Australia (CERA), University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Sergei Y Astakhov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Eugeny L Akopov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Anton Emelyanov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia.,St. Petersburg Academic University, St. Petersburg, Russia
| | | | | | | | - Saleh A Al-Obeidan
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ohoud Owaidhah
- King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | | | - Balram Chowbay
- Clinical Pharmacology, SingHealth, Singapore.,Clinical Pharmacology Laboratory, National Cancer Centre, Singapore.,Office of Clinical Sciences, Duke-NUS Medical School, Singapore
| | - Jia Nee Foo
- Genome Institute of Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | | | | | | | - Shi Qi Mok
- Genome Institute of Singapore, Singapore
| | | | | | - Su Qin Peh
- Genome Institute of Singapore, Singapore
| | | | | | - Su-Ling Ho
- Department of Ophthalmology, Tan Tock Seng Hospital, Singapore
| | | | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Academic Clinical Program for Ophthalmology and Visual Sciences, Office of Clinical and Academic Faculty Affairs, Duke-NUS Graduate Medical School, Singapore
| | | | - Rogelio González-Sarmiento
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Frederico Martinon-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain.,GENVIP Research Group, Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Spain
| | - Antonio Salas
- Unidade de Xenética, Departamento de Anatomía Patolóxica e Ciencias Forenses, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Kessara Pathanapitoon
- Department of Ophthalmology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Linda Hansapinyo
- Department of Ophthalmology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Naris Kitnarong
- Department of Ophthalmology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anavaj Sakuntabhai
- Institut Pasteur, Functional Genetics of Infectious Diseases Unit, Department of Genomes and Genetics, Paris, France.,Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Paris, France
| | - Hip X Nguyn
- Vietnam National Institute of Ophthalmology, Hanoi, Vietnam
| | | | - Trình V Nguyn
- Vietnam National Institute of Ophthalmology, Hanoi, Vietnam
| | - Werner Zenz
- Department of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Alexander Binder
- Department of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Daniela S Klobassa
- Department of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Martin L Hibberd
- Genome Institute of Singapore, Singapore.,Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Stefan Herms
- Department of Genomics, Life &Brain Center, University of Bonn, Bonn, Germany.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Division of Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Markus M Nöthen
- Department of Genomics, Life &Brain Center, University of Bonn, Bonn, Germany.,Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Robyn M Rautenbach
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Ari Ziskind
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Trevor R Carmichael
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lydia Álvarez
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain
| | - Montserrat García
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain
| | - Héctor González-Iglesias
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain
| | - Pedro P Rodríguez-Calvo
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain
| | - Luis Fernández-Vega Cueto
- Fernández-Vega University Institute and Foundation of Ophthalmological Research, University of Oviedo, Oviedo, Spain.,Fernández-Vega Ophthalmological Institute, Oviedo, Spain
| | - Çilingir Oguz
- Department of Genetics, Eskisehir Osmangazi University, Meselik, Eskisehir, Turkey
| | - Nevbahar Tamcelik
- Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Eray Atalay
- Singapore Eye Research Institute, Singapore.,Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Bilge Batu
- Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Dilek Aktas
- DAMAGEN Genetic Diagnostic Center, Ankara, Turkey
| | - Burcu Kasım
- Department of Ophthalmology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - M Roy Wilson
- School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Anne L Coleman
- Center for Community Outreach and Policy, Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Center for Biotechnology and Genomic Medicine, James and Jean Culver Discovery Institute, Augusta University, Augusta, Georgia, USA
| | - Pratap Challa
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, USA
| | - Leon Herndon
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, USA
| | - Rachel W Kuchtey
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Kuchtey
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Karen Curtin
- John A. Moran Eye Center, Department of Ophthalmology, University of Utah, Salt Lake City, Utah, USA
| | - Craig J Chaya
- John A. Moran Eye Center, Department of Ophthalmology, University of Utah, Salt Lake City, Utah, USA
| | - Alan Crandall
- John A. Moran Eye Center, Department of Ophthalmology, University of Utah, Salt Lake City, Utah, USA
| | - Linda M Zangwill
- Hamilton Glaucoma Center, Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, San Diego, California, USA
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Anneke I den Hollander
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Eija Vesti
- Department of Ophthalmology, University of Turku and Turku University Hospital, Turku, Finland
| | - John H Fingert
- Institute for Vision Research, University of Iowa, Iowa City, Iowa, USA.,Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Arthur J Sit
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China.,Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Daniele Cusi
- Institute of Biomedical Technologies, Italian National Research Centre (ITB-CNR), Segrate-Milano, Italy
| | - Raheel Qamar
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan.,Department of Biochemistry, Al-Nafees Medical College and Hospital, Isra University, Islamabad, Pakistan
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Soumya Raychaudhuri
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Institute of Inflammation and Repair, University of Manchester, Manchester, UK.,Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Pathology, Rigshospitalet, Eye Pathology Section, University of Copenhagen, Copenhagen, Denmark
| | - Tero Kivelä
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - André Reis
- David Tvildiani Medical University, Tbilisi, Georgia
| | - Friedrich E Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, San Diego, California, USA
| | - Louis R Pasquale
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,Institute of Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Fridbert Jonasson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Ophthalmology, Landspitali University Hospital, Reykjavik, Iceland
| | - R Rand Allingham
- Singapore Eye Research Institute, Singapore.,Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, USA
| | - Dan Milea
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Academic Clinical Program for Ophthalmology and Visual Sciences, Office of Clinical and Academic Faculty Affairs, Duke-NUS Graduate Medical School, Singapore
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Toshiaki Kubota
- Department of Ophthalmology, Oita University Faculty of Medicine, Oita, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eranga N Vithana
- Singapore Eye Research Institute, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazia Micheal
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Fotis Topouzis
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Michael Dubina
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia.,St. Petersburg Academic University, St. Petersburg, Russia
| | - Periasamy Sundaresan
- Dr. G.Venkataswamy Eye Research Institute, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, India
| | - Kari Stefansson
- deCODE Genetics, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Francesca Pasutto
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Chiea Chuen Khor
- Singapore Eye Research Institute, Singapore.,Genome Institute of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
255
|
Lau W, Andrew T, Maniatis N. High-Resolution Genetic Maps Identify Multiple Type 2 Diabetes Loci at Regulatory Hotspots in African Americans and Europeans. Am J Hum Genet 2017; 100:803-816. [PMID: 28475862 DOI: 10.1016/j.ajhg.2017.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/11/2017] [Indexed: 10/19/2022] Open
Abstract
Interpretation of results from genome-wide association studies for T2D is challenging. Only very few loci have been replicated in African ancestry populations and the identification of the implicated functional genes remain largely undefined. We used genetic maps that capture detailed linkage disequilibrium information in European and African Americans and applied these to large T2D case-control samples in order to estimate locations for putative functional variants in both populations. Replicated T2D locations were tested for evidence of being regulatory hotspots using adipose expression. We validated a sample of our co-location intervals using next generation sequencing and functional annotation, including enhancers, transcription, and chromatin modifications. We identified 111 additional disease-susceptibility locations, 93 of which are cosmopolitan and 18 of which are European specific. We show that many previously known signals are also risk loci in African Americans. The majority of the disease locations appear to confer risk of T2D via the regulation of expression levels for a large number (266) of cis-regulated genes, the majority of which are not the nearest genes to the disease loci. Sequencing three cosmopolitan locations provided candidate functional variants that precisely co-locate with cell-specific chromatin domains and pancreatic islet enhancers. These variants have large effect sizes and are common across populations. Results show that disease-associated loci in different populations, gene expression, and cell-specific regulatory annotation can be effectively integrated by localizing these effects on high-resolution genetic maps. The cis-regulated genes provide insights into the complex molecular pathways involved and can be used as targets for sequencing and functional molecular studies.
Collapse
|
256
|
Abstract
The current focus on delivery of personalised (or precision) medicine reflects the expectation that developments in genomics, imaging and other domains will extend our diagnostic and prognostic capabilities, and enable more effective targeting of current and future preventative and therapeutic options. The clinical benefits of this approach are already being realised in rare diseases and cancer but the impact on management of complex diseases, such as type 2 diabetes, remains limited. This may reflect reliance on inappropriate models of disease architecture, based around rare, high-impact genetic and environmental exposures that are poorly suited to our emerging understanding of type 2 diabetes. This review proposes an alternative 'palette' model, centred on a molecular taxonomy that focuses on positioning an individual with respect to the major pathophysiological processes that contribute to diabetes risk and progression. This model anticipates that many individuals with diabetes will have multiple parallel defects that affect several of these processes. One corollary of this model is that research efforts should, at least initially, be targeted towards identifying and characterising individuals whose adverse metabolic trajectory is dominated by perturbation in a restricted set of processes.
Collapse
Affiliation(s)
- Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK.
| |
Collapse
|
257
|
Mulder H. Melatonin signalling and type 2 diabetes risk: too little, too much or just right? Diabetologia 2017; 60:826-829. [PMID: 28303303 DOI: 10.1007/s00125-017-4249-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/06/2017] [Indexed: 01/29/2023]
Abstract
Of the associations of genetic variants with type 2 diabetes, the one of an SNP in an intron of the gene encoding the melatonin receptor 1B (MTNR1B) has been remarkably robust. Work from our group and others has provided support for a model where carriers of this risk G allele exhibit increased MTNR1B expression in islets of Langerhans. Most published studies to date favour that melatonin's action on the beta cell is inhibition of insulin secretion. Hence, our model proposes that this inhibitory effect of melatonin is exaggerated in carriers of the MTNR1B risk G allele. This would explain why this genetic association causes reduced insulin secretion and greater risk of future type 2 diabetes, as has been observed in numerous studies. Concurrently, another body of work has shown that rare MTNR1B alleles, which could perturb receptor function, also associate with type 2 diabetes. In this commentary, it is suggested that such apparently conflicting observations can be reconciled by the fact that non-coding (intronic; frequent) and coding (exonic; rare) alleles of MTNR1B give rise to different phenotypes. Thus, altered gene transcription may explain why SNPs, which do not alter coding sequences, exhibit cell-specific effects. In contrast, SNPs that change protein sequences are more likely to exert generalised effects since an altered protein will appear in all cells expressing the gene.
Collapse
Affiliation(s)
- Hindrik Mulder
- Unit of Molecular Metabolism, Lund University Diabetes Centre, Jan Waldenströms gata 35, SE-205 02, Malmö, Sweden.
| |
Collapse
|
258
|
Lopez-Minguez J, Saxena R, Bandín C, Scheer FA, Garaulet M. Late dinner impairs glucose tolerance in MTNR1B risk allele carriers: A randomized, cross-over study. Clin Nutr 2017; 37:1133-1140. [PMID: 28455106 DOI: 10.1016/j.clnu.2017.04.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Late-night dinner eating is associated with increased risk for type-2 diabetes. The underlying mechanism is unclear. One explanatory hypothesis is that the concurrence of elevated circulating melatonin and high glucose concentrations (characterizing late eating) leads to impaired glucose tolerance. However, to date no study has tested the influence of physiological melatonin concentrations on glucose-tolerance. The discovery of melatonin receptor MTNR1B as a diabetes risk gene provides evidence for a role of physiological levels of melatonin in glucose control. The aim of our study was to test the hypothesis that elevated endogenous melatonin concentrations worsen glucose control when eating late. Registered under ClinicalTrials.gov Identifier no. NCT03003936. METHODS We performed a randomized, cross-over trial to compare glucose tolerance in the presence (late dinner) or absence (early dinner) of elevated physiological melatonin concentrations and we compared the results between homozygous carriers and non-carriers of the MTNR1B risk allele. RESULTS The concurrence of meal timing with elevated endogenous melatonin concentrations resulted in impaired glucose tolerance. This effect was stronger in MTNR1B risk-carriers than in non-carriers. Furthermore, eating late significantly impaired glucose tolerance only in risk-carriers and not in the non-risk carriers. CONCLUSIONS The interaction of dinner timing with MTNR1B supports a causal role of endogenous melatonin in the impairment of glucose tolerance. These results suggest that moving the dinner to an earlier time may result in better glucose tolerance specially in MTNR1B carriers. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT03003936.
Collapse
Affiliation(s)
- Jesus Lopez-Minguez
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Broad Institute, Cambridge, MA, USA
| | - Cristina Bandín
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain
| | - Frank A Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain.
| |
Collapse
|
259
|
Martin AR, Gignoux CR, Walters RK, Wojcik GL, Neale BM, Gravel S, Daly MJ, Bustamante CD, Kenny EE. Human Demographic History Impacts Genetic Risk Prediction across Diverse Populations. Am J Hum Genet 2017. [PMID: 28366442 DOI: 10.1016/j.ajhg] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
The vast majority of genome-wide association studies (GWASs) are performed in Europeans, and their transferability to other populations is dependent on many factors (e.g., linkage disequilibrium, allele frequencies, genetic architecture). As medical genomics studies become increasingly large and diverse, gaining insights into population history and consequently the transferability of disease risk measurement is critical. Here, we disentangle recent population history in the widely used 1000 Genomes Project reference panel, with an emphasis on populations underrepresented in medical studies. To examine the transferability of single-ancestry GWASs, we used published summary statistics to calculate polygenic risk scores for eight well-studied phenotypes. We identify directional inconsistencies in all scores; for example, height is predicted to decrease with genetic distance from Europeans, despite robust anthropological evidence that West Africans are as tall as Europeans on average. To gain deeper quantitative insights into GWAS transferability, we developed a complex trait coalescent-based simulation framework considering effects of polygenicity, causal allele frequency divergence, and heritability. As expected, correlations between true and inferred risk are typically highest in the population from which summary statistics were derived. We demonstrate that scores inferred from European GWASs are biased by genetic drift in other populations even when choosing the same causal variants and that biases in any direction are possible and unpredictable. This work cautions that summarizing findings from large-scale GWASs may have limited portability to other populations using standard approaches and highlights the need for generalized risk prediction methods and the inclusion of more diverse individuals in medical genomics.
Collapse
Affiliation(s)
- Alicia R Martin
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Simon Gravel
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada; McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Eimear E Kenny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center of Statistical Genetics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
260
|
Human Demographic History Impacts Genetic Risk Prediction across Diverse Populations. Am J Hum Genet 2017; 100:635-649. [PMID: 28366442 DOI: 10.1016/j.ajhg.2017.03.004] [Citation(s) in RCA: 891] [Impact Index Per Article: 111.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/10/2017] [Indexed: 01/10/2023] Open
Abstract
The vast majority of genome-wide association studies (GWASs) are performed in Europeans, and their transferability to other populations is dependent on many factors (e.g., linkage disequilibrium, allele frequencies, genetic architecture). As medical genomics studies become increasingly large and diverse, gaining insights into population history and consequently the transferability of disease risk measurement is critical. Here, we disentangle recent population history in the widely used 1000 Genomes Project reference panel, with an emphasis on populations underrepresented in medical studies. To examine the transferability of single-ancestry GWASs, we used published summary statistics to calculate polygenic risk scores for eight well-studied phenotypes. We identify directional inconsistencies in all scores; for example, height is predicted to decrease with genetic distance from Europeans, despite robust anthropological evidence that West Africans are as tall as Europeans on average. To gain deeper quantitative insights into GWAS transferability, we developed a complex trait coalescent-based simulation framework considering effects of polygenicity, causal allele frequency divergence, and heritability. As expected, correlations between true and inferred risk are typically highest in the population from which summary statistics were derived. We demonstrate that scores inferred from European GWASs are biased by genetic drift in other populations even when choosing the same causal variants and that biases in any direction are possible and unpredictable. This work cautions that summarizing findings from large-scale GWASs may have limited portability to other populations using standard approaches and highlights the need for generalized risk prediction methods and the inclusion of more diverse individuals in medical genomics.
Collapse
|
261
|
Cheng M, Liu X, Yang M, Han L, Xu A, Huang Q. Computational analyses of type 2 diabetes-associated loci identified by genome-wide association studies. J Diabetes 2017; 9:362-377. [PMID: 27121852 DOI: 10.1111/1753-0407.12421] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/31/2016] [Accepted: 04/23/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) of type 2 diabetes (T2D) have discovered a number of loci that contribute to susceptibility to the disease. Future challenges include elucidation of functional mechanisms through which these GWAS-identified loci modulate T2D disease risk. The aim of the present study was to comprehensively characterize T2D associated single nucleotide polymorphisms (SNPs) and genes through computational approaches. METHODS Computational biology approaches used in the present study included comparative genomic analyses and functional annotation using GWAS3D and RegulomeDB, investigation of the effects of T2D-associated SNPs on miRNA binding and protein phosphorylation, and gene ontology, pathway enrichment, protein-protein interaction (PPI) networks and functional module analysis of T2D-associated genes from previously published GWAS. RESULTS Computational analysis identified a number of T2D GWAS-associated SNPs that were located at protein binding sites, including CCCTC-binding factor (CTCF), E1A binding protein p300 (EP300), hepatocyte nuclear factor 4alpha (HNF4A), transcription factor 7 like 2 (TCF7L2), forkhead box A1 (FOXA1) and A2 (FOXA2), and potentially affected the binding of miRNAs and protein phosphorylation. Pathway enrichment analysis confirmed two well-known maturity onset diabetes of the young and T2D pathways, whereas PPI network analysis identified highly interconnected "hub" genes, such as TCF7L2, melatonin receptor 1B (MTNR1B), and solute carrier family 30 (zinc transporter), member 8 (SLC30A8), that created two tight subnetworks. CONCLUSIONS The results provide objectives and clues for future experimental studies and further insights into the molecular pathogenesis of T2D.
Collapse
Affiliation(s)
- Mengrong Cheng
- College of Life Sciences, Central China Normal University, Wuhan, China
| | - Xinhong Liu
- College of Life Sciences, Central China Normal University, Wuhan, China
| | - Mei Yang
- College of Life Sciences, Central China Normal University, Wuhan, China
| | - Lanchun Han
- College of Life Sciences, Central China Normal University, Wuhan, China
- Institute of Public Health and Molecular Medicine Analysis, Central China Normal University, Wuhan, China
| | - Aimin Xu
- Li Cha Chung Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qingyang Huang
- College of Life Sciences, Central China Normal University, Wuhan, China
- Institute of Public Health and Molecular Medicine Analysis, Central China Normal University, Wuhan, China
| |
Collapse
|
262
|
Bastidas-Ponce A, Roscioni SS, Burtscher I, Bader E, Sterr M, Bakhti M, Lickert H. Foxa2 and Pdx1 cooperatively regulate postnatal maturation of pancreatic β-cells. Mol Metab 2017; 6:524-534. [PMID: 28580283 PMCID: PMC5444078 DOI: 10.1016/j.molmet.2017.03.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The transcription factors (TF) Foxa2 and Pdx1 are key regulators of beta-cell (β-cell) development and function. Mutations of these TFs or their respective cis-regulatory consensus binding sites have been linked to maturity diabetes of the young (MODY), pancreas agenesis, or diabetes susceptibility in human. Although Foxa2 has been shown to directly regulate Pdx1 expression during mouse embryonic development, the impact of this gene regulatory interaction on postnatal β-cell maturation remains obscure. METHODS In order to easily monitor the expression domains of Foxa2 and Pdx1 and analyze their functional interconnection, we generated a novel double knock-in homozygous (FVFPBFDHom) fluorescent reporter mouse model by crossing the previously described Foxa2-Venus fusion (FVF) with the newly generated Pdx1-BFP (blue fluorescent protein) fusion (PBF) mice. RESULTS Although adult PBF homozygous animals exhibited a reduction in expression levels of Pdx1, they are normoglycemic. On the contrary, despite normal pancreas and endocrine development, the FVFPBFDHom reporter male animals developed hyperglycemia at weaning age and displayed a reduction in Pdx1 levels in islets, which coincided with alterations in β-cell number and islet architecture. The failure to establish mature β-cells resulted in loss of β-cell identity and trans-differentiation towards other endocrine cell fates. Further analysis suggested that Foxa2 and Pdx1 genetically and functionally cooperate to regulate maturation of adult β-cells. CONCLUSIONS Our data show that the maturation of pancreatic β-cells requires the cooperative function of Foxa2 and Pdx1. Understanding the postnatal gene regulatory network of β-cell maturation will help to decipher pathomechanisms of diabetes and identify triggers to regenerate dedifferentiated β-cell mass.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,German Center for Diabetes Research (DZD), Germany
| | - Sara S Roscioni
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,German Center for Diabetes Research (DZD), Germany
| | - Erik Bader
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,German Center for Diabetes Research (DZD), Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,Technical University of Munich, Germany.,German Center for Diabetes Research (DZD), Germany
| |
Collapse
|
263
|
Zhao Z, Li X, Jian D, Hao P, Rao L, Li M. Hsa_circ_0054633 in peripheral blood can be used as a diagnostic biomarker of pre-diabetes and type 2 diabetes mellitus. Acta Diabetol 2017; 54:237-245. [PMID: 27878383 PMCID: PMC5329094 DOI: 10.1007/s00592-016-0943-0] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022]
Abstract
AIMS The purpose of the current study was to investigate the characteristic expression of circular RNAs (circRNAs) in the peripheral blood of type 2 diabetes mellitus (T2DM) patients and their potential as diagnostic biomarkers for pre-diabetes and T2DM. METHODS CircRNAs in the peripheral blood from six healthy individuals and six T2DM patients were collected for microarray analysis, and an independent cohort study consisting of 20 normal cases, 20 pre-diabetes patients and 20 T2DM patients was conducted to verify the five chosen circRNAs. We then tested hsa_circ_0054633 in a third cohort (control group, n = 60; pre-diabetes group, n = 63; and T2DM group, n = 64) by quantitative real-time polymerase chain reaction (Q-PCR). RESULTS In total, 489 circRNAs were discovered to be differentially expressed between the two groups, and of these, 78 were upregulated and 411 were downregulated in the T2DM group. Five circRNAs were then selected as candidate biomarkers and further verified in a second cohort. Hsa_circ_0054633 was found to have the largest area under the curve (AUC). The diagnostic capacity of hsa_circ_0054633 was tested in a third cohort. After introducing the risk factors of T2DM, the hsa_circ_0054633 AUCs for the diagnosis of pre-diabetes and T2DM slightly increased from 0.751 (95% confidence interval [0.666-0.835], P < 0.001) to 0.841 ([0.773-0.910], P < 0.001) and from 0.793 ([0.716-0.871], P < 0.001) to 0.834 ([0.762-0.905], P < 0.001), respectively. CONCLUSIONS Hsa_circ_0054633 presented a certain diagnostic capability for pre-diabetes and T2DM.
Collapse
Affiliation(s)
- Zhenzhou Zhao
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xuejie Li
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Dongdong Jian
- Department of Cardiology, The First Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, China
| | - Peiyuan Hao
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lixin Rao
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Muwei Li
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
264
|
Pulit SL, Karaderi T, Lindgren CM. Sexual dimorphisms in genetic loci linked to body fat distribution. Biosci Rep 2017; 37:BSR20160184. [PMID: 28073971 PMCID: PMC5291139 DOI: 10.1042/bsr20160184] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/07/2017] [Accepted: 01/10/2017] [Indexed: 01/02/2023] Open
Abstract
Obesity is a chronic condition associated with increased morbidity and mortality and is a risk factor for a number of other diseases including type 2 diabetes and cardiovascular disease. Obesity confers an enormous, costly burden on both individuals and public health more broadly. Body fat distribution is a heritable trait and a well-established predictor of adverse metabolic outcomes. Body fat distribution is distinct from overall obesity in measurement, but studies of body fat distribution can yield insights into the risk factors for and causes of overall obesity. Sexual dimorphism in body fat distribution is present throughout life. Though sexual dimorphism is subtle in early stages of life, it is attenuated in puberty and during menopause. This phenomenon could be, at least in part, due to the influence of sex hormones on the trait. Findings from recent large genome-wide association studies (GWAS) for various measures of body fat distribution (including waist-to-hip ratio, hip or waist circumference, trunk fat percentage and the ratio of android and gynoid fat percentage) emphasize the strong sexual dimorphism in the genetic regulation of fat distribution traits. Importantly, sexual dimorphism is not observed for overall obesity (as assessed by body mass index or total fat percentage). Notably, the genetic loci associated with body fat distribution, which show sexual dimorphism, are located near genes that are expressed in adipose tissues and/or adipose cells. Considering the epidemiological and genetic evidence, sexual dimorphism is a prominent feature of body fat distribution. Research that specifically focuses on sexual dimorphism in fat distribution can provide novel insights into human physiology and into the development of obesity and its comorbidities, as well as yield biological clues that will aid in the improvement of disease prevention and treatment.
Collapse
Affiliation(s)
- Sara L Pulit
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tugce Karaderi
- Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, U.K.
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| |
Collapse
|
265
|
Lawlor N, Khetan S, Ucar D, Stitzel ML. Genomics of Islet (Dys)function and Type 2 Diabetes. Trends Genet 2017; 33:244-255. [PMID: 28245910 DOI: 10.1016/j.tig.2017.01.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/30/2017] [Indexed: 12/28/2022]
Abstract
Pancreatic islet dysfunction and beta cell failure are hallmarks of type 2 diabetes mellitus (T2DM) pathogenesis. In this review, we discuss how genome-wide association studies (GWASs) and recent developments in islet (epi)genome and transcriptome profiling (particularly single cell analyses) are providing novel insights into the genetic, environmental, and cellular contributions to islet (dys)function and T2DM pathogenesis. Moving forward, study designs that interrogate and model genetic variation [e.g., allelic profiling and (epi)genome editing] will be critical to dissect the molecular genetics of T2DM pathogenesis, to build next-generation cellular and animal models, and to develop precision medicine approaches to detect, treat, and prevent islet (dys)function and T2DM.
Collapse
Affiliation(s)
- Nathan Lawlor
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Shubham Khetan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT 06032, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT 06032, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
266
|
Genetic regulatory signatures underlying islet gene expression and type 2 diabetes. Proc Natl Acad Sci U S A 2017; 114:2301-2306. [PMID: 28193859 DOI: 10.1073/pnas.1621192114] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified >100 independent SNPs that modulate the risk of type 2 diabetes (T2D) and related traits. However, the pathogenic mechanisms of most of these SNPs remain elusive. Here, we examined genomic, epigenomic, and transcriptomic profiles in human pancreatic islets to understand the links between genetic variation, chromatin landscape, and gene expression in the context of T2D. We first integrated genome and transcriptome variation across 112 islet samples to produce dense cis-expression quantitative trait loci (cis-eQTL) maps. Additional integration with chromatin-state maps for islets and other diverse tissue types revealed that cis-eQTLs for islet-specific genes are specifically and significantly enriched in islet stretch enhancers. High-resolution chromatin accessibility profiling using assay for transposase-accessible chromatin sequencing (ATAC-seq) in two islet samples enabled us to identify specific transcription factor (TF) footprints embedded in active regulatory elements, which are highly enriched for islet cis-eQTL. Aggregate allelic bias signatures in TF footprints enabled us de novo to reconstruct TF binding affinities genetically, which support the high-quality nature of the TF footprint predictions. Interestingly, we found that T2D GWAS loci were strikingly and specifically enriched in islet Regulatory Factor X (RFX) footprints. Remarkably, within and across independent loci, T2D risk alleles that overlap with RFX footprints uniformly disrupt the RFX motifs at high-information content positions. Together, these results suggest that common regulatory variations have shaped islet TF footprints and the transcriptome and that a confluent RFX regulatory grammar plays a significant role in the genetic component of T2D predisposition.
Collapse
|
267
|
Decreased STARD10 Expression Is Associated with Defective Insulin Secretion in Humans and Mice. Am J Hum Genet 2017; 100:238-256. [PMID: 28132686 PMCID: PMC5294761 DOI: 10.1016/j.ajhg.2017.01.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/20/2016] [Indexed: 12/30/2022] Open
Abstract
Genetic variants near ARAP1 (CENTD2) and STARD10 influence type 2 diabetes (T2D) risk. The risk alleles impair glucose-induced insulin secretion and, paradoxically but characteristically, are associated with decreased proinsulin:insulin ratios, indicating improved proinsulin conversion. Neither the identity of the causal variants nor the gene(s) through which risk is conferred have been firmly established. Whereas ARAP1 encodes a GTPase activating protein, STARD10 is a member of the steroidogenic acute regulatory protein (StAR)-related lipid transfer protein family. By integrating genetic fine-mapping and epigenomic annotation data and performing promoter-reporter and chromatin conformational capture (3C) studies in β cell lines, we localize the causal variant(s) at this locus to a 5 kb region that overlaps a stretch-enhancer active in islets. This region contains several highly correlated T2D-risk variants, including the rs140130268 indel. Expression QTL analysis of islet transcriptomes from three independent subject groups demonstrated that T2D-risk allele carriers displayed reduced levels of STARD10 mRNA, with no concomitant change in ARAP1 mRNA levels. Correspondingly, β-cell-selective deletion of StarD10 in mice led to impaired glucose-stimulated Ca2+ dynamics and insulin secretion and recapitulated the pattern of improved proinsulin processing observed at the human GWAS signal. Conversely, overexpression of StarD10 in the adult β cell improved glucose tolerance in high fat-fed animals. In contrast, manipulation of Arap1 in β cells had no impact on insulin secretion or proinsulin conversion in mice. This convergence of human and murine data provides compelling evidence that the T2D risk associated with variation at this locus is mediated through reduction in STARD10 expression in the β cell.
Collapse
|
268
|
Najmi LA, Aukrust I, Flannick J, Molnes J, Burtt N, Molven A, Groop L, Altshuler D, Johansson S, Bjørkhaug L, Njølstad PR. Functional Investigations of HNF1A Identify Rare Variants as Risk Factors for Type 2 Diabetes in the General Population. Diabetes 2017; 66:335-346. [PMID: 27899486 PMCID: PMC5860263 DOI: 10.2337/db16-0460] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/18/2016] [Indexed: 12/18/2022]
Abstract
Variants in HNF1A encoding hepatocyte nuclear factor 1α (HNF-1A) are associated with maturity-onset diabetes of the young form 3 (MODY 3) and type 2 diabetes. We investigated whether functional classification of HNF1A rare coding variants can inform models of diabetes risk prediction in the general population by analyzing the effect of 27 HNF1A variants identified in well-phenotyped populations (n = 4,115). Bioinformatics tools classified 11 variants as likely pathogenic and showed no association with diabetes risk (combined minor allele frequency [MAF] 0.22%; odds ratio [OR] 2.02; 95% CI 0.73-5.60; P = 0.18). However, a different set of 11 variants that reduced HNF-1A transcriptional activity to <60% of normal (wild-type) activity was strongly associated with diabetes in the general population (combined MAF 0.22%; OR 5.04; 95% CI 1.99-12.80; P = 0.0007). Our functional investigations indicate that 0.44% of the population carry HNF1A variants that result in a substantially increased risk for developing diabetes. These results suggest that functional characterization of variants within MODY genes may overcome the limitations of bioinformatics tools for the purposes of presymptomatic diabetes risk prediction in the general population.
Collapse
Affiliation(s)
- Laeya Abdoli Najmi
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ingvild Aukrust
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Janne Molnes
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Noel Burtt
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Anders Molven
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Clinical Research Center, Lund University, Malmö, Sweden
| | - David Altshuler
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
- Departments of Genetics and Medicine, Harvard Medical School, Boston, MA
- Departments of Molecular Biology and Diabetes Unit, Massachusetts General Hospital, Boston, MA
| | - Stefan Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lise Bjørkhaug
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Biomedical Laboratory Sciences, Bergen University College, Bergen, Norway
| | - Pål Rasmus Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
269
|
Skyler JS, Bakris GL, Bonifacio E, Darsow T, Eckel RH, Groop L, Groop PH, Handelsman Y, Insel RA, Mathieu C, McElvaine AT, Palmer JP, Pugliese A, Schatz DA, Sosenko JM, Wilding JPH, Ratner RE. Differentiation of Diabetes by Pathophysiology, Natural History, and Prognosis. Diabetes 2017; 66:241-255. [PMID: 27980006 PMCID: PMC5384660 DOI: 10.2337/db16-0806] [Citation(s) in RCA: 411] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022]
Abstract
The American Diabetes Association, JDRF, the European Association for the Study of Diabetes, and the American Association of Clinical Endocrinologists convened a research symposium, "The Differentiation of Diabetes by Pathophysiology, Natural History and Prognosis" on 10-12 October 2015. International experts in genetics, immunology, metabolism, endocrinology, and systems biology discussed genetic and environmental determinants of type 1 and type 2 diabetes risk and progression, as well as complications. The participants debated how to determine appropriate therapeutic approaches based on disease pathophysiology and stage and defined remaining research gaps hindering a personalized medical approach for diabetes to drive the field to address these gaps. The authors recommend a structure for data stratification to define the phenotypes and genotypes of subtypes of diabetes that will facilitate individualized treatment.
Collapse
Affiliation(s)
- Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | | | | | - Robert H Eckel
- University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Leif Groop
- Lund University, Skåne University Hospital, Malmö, Sweden
| | - Per-Henrik Groop
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | | | | | | | - Jerry P Palmer
- University of Washington and VA Puget Sound Health Care System, Seattle, WA
| | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | - Jay M Sosenko
- University of Miami Miller School of Medicine, Miami, FL
| | | | | |
Collapse
|
270
|
Affiliation(s)
- Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Old Road, Headington, Oxford OX3 7LJ, UK; and at the Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7BN, UK
| |
Collapse
|
271
|
Laakso M, Kuusisto J, Stančáková A, Kuulasmaa T, Pajukanta P, Lusis AJ, Collins FS, Mohlke KL, Boehnke M. The Metabolic Syndrome in Men study: a resource for studies of metabolic and cardiovascular diseases. J Lipid Res 2017; 58:481-493. [PMID: 28119442 DOI: 10.1194/jlr.o072629] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/15/2017] [Indexed: 12/30/2022] Open
Abstract
The Metabolic Syndrome in Men (METSIM) study is a population-based study including 10,197 Finnish men examined in 2005-2010. The aim of the study is to investigate nongenetic and genetic factors associated with the risk of T2D and CVD, and with cardiovascular risk factors. The protocol includes a detailed phenotyping of the participants, an oral glucose tolerance test, fasting laboratory measurements including proton NMR measurements, mass spectometry metabolomics, adipose tissue biopsies from 1,400 participants, and a stool sample. In our ongoing follow-up study, we have, to date, reexamined 6,496 participants. Extensive genotyping and exome sequencing have been performed for essentially all METSIM participants, and >2,000 METSIM participants have been whole-genome sequenced. We have identified several nongenetic markers associated with the development of diabetes and cardiovascular events, and participated in several genetic association studies to identify gene variants associated with diabetes, hyperglycemia, and cardiovascular risk factors. The generation of a phenotype and genotype resource in the METSIM study allows us to proceed toward a "systems genetics" approach, which includes statistical methods to quantitate and integrate intermediate phenotypes, such as transcript, protein, or metabolite levels, to provide a global view of the molecular architecture of complex traits.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland .,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Aldons J Lusis
- Departments of Human Genetics David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA.,Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
272
|
Zhu M, Geng L, Shen W, Wang Y, Liu J, Cheng Y, Wang C, Dai J, Jin G, Hu Z, Ma H, Shen H. Exome-Wide Association Study Identifies Low-Frequency Coding Variants in 2p23.2 and 7p11.2 Associated with Survival of Non-Small Cell Lung Cancer Patients. J Thorac Oncol 2017; 12:644-656. [PMID: 28104536 DOI: 10.1016/j.jtho.2016.12.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/23/2016] [Accepted: 12/15/2016] [Indexed: 01/10/2023]
Abstract
INTRODUCTION A growing body of evidence has suggested that low-frequency or rare coding variants might have strong effects on the development and prognosis of cancer. Here, we aim to assess the role of low-frequency and rare coding variants in the survival of NSCLC in Chinese populations. METHODS We performed an exome-wide scan of 247,870 variants in 1008 patients with NSCLC and replicated the promising variants by using imputed genotype data of The Cancer Genome Atlas (TCGA) with a Cox regression model. Gene-based and pathway-based analysis were also performed for nonsynonymous or splice site variants. Additionally, analysis of gene expression data in the TCGA was used to increase the reliability of candidate loci and genes. RESULTS A low-frequency missense variant in chaperonin containing TCP1 subunit 6A gene (CCT6A) (rs33922584: adjusted hazard ratio [HRadjusted] = 1.75, p = 6.06 × 10-4) was significantly related to the survival of patients with NSCLC, which was further replicated by the TCGA samples (HRadjusted = 4.19, p = 0.015). Interestingly, the G allele of rs33922584 was significantly associated with high expression of CCT6A (p = 0.019) that might induce the worse survival in the TCGA samples (HRadjusted = 1.15, p = 0.047). Besides, rs117512489 in gene phospholipase B1 gene (PLB1) (HR = 2.02, p = 7.28 × 10-4) was also associated with survival of the patients with NSCLC in our samples, but it was supported only by gene expression analysis in the TCGA (HRadjusted = 1.15, p = 0.023). Gene-based and pathway-based analysis revealed a total of 32 genes, including CCT6A and 34 potential pathways might account for the survival of NSCLC, respectively. CONCLUSION These results provided more evidence for the important role of low-frequency or rare variants in the survival of patients with NSCLC.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Liguo Geng
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Shen
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yuzhuo Wang
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jia Liu
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yang Cheng
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University, Nanjing, People's Republic of China.
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center of Cancer Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
273
|
Rosta K, Al-Aissa Z, Hadarits O, Harreiter J, Nádasdi Á, Kelemen F, Bancher-Todesca D, Komlósi Z, Németh L, Rigó J, Sziller I, Somogyi A, Kautzky-Willer A, Firneisz G. Association Study with 77 SNPs Confirms the Robust Role for the rs10830963/G of MTNR1B Variant and Identifies Two Novel Associations in Gestational Diabetes Mellitus Development. PLoS One 2017; 12:e0169781. [PMID: 28072873 PMCID: PMC5224877 DOI: 10.1371/journal.pone.0169781] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022] Open
Abstract
CONTEXT Genetic variation in human maternal DNA contributes to the susceptibility for development of gestational diabetes mellitus (GDM). OBJECTIVE We assessed 77 maternal single nucleotide gene polymorphisms (SNPs) for associations with GDM or plasma glucose levels at OGTT in pregnancy. METHODS 960 pregnant women (after dropouts 820: case/control: m99'WHO: 303/517, IADPSG: 287/533) were enrolled in two countries into this case-control study. After genomic DNA isolation the 820 samples were collected in a GDM biobank and assessed using KASP (LGC Genomics) genotyping assay. Logistic regression risk models were used to calculate ORs according to IADPSG/m'99WHO criteria based on standard OGTT values. RESULTS The most important risk alleles associated with GDM were rs10830963/G of MTNR1B (OR = 1.84/1.64 [IADPSG/m'99WHO], p = 0.0007/0.006), rs7754840/C (OR = 1.51/NS, p = 0.016) of CDKAL1 and rs1799884/T (OR = 1.4/1.56, p = 0.04/0.006) of GCK. The rs13266634/T (SLC30A8, OR = 0.74/0.71, p = 0.05/0.02) and rs7578326/G (LOC646736/IRS1, OR = 0.62/0.60, p = 0.001/0.006) variants were associated with lower risk to develop GDM. Carrying a minor allele of rs10830963 (MTNR1B); rs7903146 (TCF7L2); rs1799884 (GCK) SNPs were associated with increased plasma glucose levels at routine OGTT. CONCLUSIONS We confirmed the robust association of MTNR1B rs10830963/G variant with GDM binary and glycemic traits in this Caucasian case-control study. As novel associations we report the minor, G allele of the rs7578326 SNP in the LOC646736/IRS1 region as a significant and the rs13266634/T SNP (SLC30A8) as a suggestive protective variant against GDM development. Genetic susceptibility appears to be more preponderant in individuals who meet both the modified 99'WHO and the IADPSG GDM diagnostic criteria.
Collapse
Affiliation(s)
- Klara Rosta
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Zahra Al-Aissa
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Orsolya Hadarits
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Jürgen Harreiter
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ákos Nádasdi
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Fanni Kelemen
- University of Szeged, Faculty of Medicine, Szeged, Hungary
| | | | - Zsolt Komlósi
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - László Németh
- Department of Probability Theory and Statistics, Eötvös Loránd University, Budapest, Hungary
| | - János Rigó
- 1 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - István Sziller
- Department of Obstetrics and Gynecology, Szent Imre Teaching Hospital, Budapest, Hungary
| | - Anikó Somogyi
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gábor Firneisz
- 2 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Academy of Sciences - Semmelweis University, Molecular Medicine Research Group, Budapest, Hungary
- * E-mail:
| |
Collapse
|
274
|
Hizel C, Tremblay J, Bartlett G, Hamet P. Introduction. PROGRESS AND CHALLENGES IN PRECISION MEDICINE 2017:1-34. [DOI: 10.1016/b978-0-12-809411-2.00001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
275
|
The InterAct Consortium. Investigation of gene-diet interactions in the incretin system and risk of type 2 diabetes: the EPIC-InterAct study. Diabetologia 2016; 59:2613-2621. [PMID: 27623947 PMCID: PMC6518069 DOI: 10.1007/s00125-016-4090-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/18/2016] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS The gut incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) have a major role in the pathophysiology of type 2 diabetes. Specific genetic and dietary factors have been found to influence the release and action of incretins. We examined the effect of interactions between seven incretin-related genetic variants in GIPR, KCNQ1, TCF7L2 and WFS1 and dietary components (whey-containing dairy, cereal fibre, coffee and olive oil) on the risk of type 2 diabetes in the European Prospective Investigation into Cancer and Nutrition (EPIC)-InterAct study. METHODS The current case-cohort study included 8086 incident type 2 diabetes cases and a representative subcohort of 11,035 participants (median follow-up: 12.5 years). Prentice-weighted Cox proportional hazard regression models were used to investigate the associations and interactions between the dietary factors and genes in relation to the risk of type 2 diabetes. RESULTS An interaction (p = 0.048) between TCF7L2 variants and coffee intake was apparent, with an inverse association between coffee and type 2 diabetes present among carriers of the diabetes risk allele (T) in rs12255372 (GG: HR 0.99 [95% CI 0.97, 1.02] per cup of coffee; GT: HR 0.96 [95% CI 0.93, 0.98]); and TT: HR 0.93 [95% CI 0.88, 0.98]). In addition, an interaction (p = 0.005) between an incretin-specific genetic risk score and coffee was observed, again with a stronger inverse association with coffee in carriers with more risk alleles (0-3 risk alleles: HR 0.99 [95% CI 0.94, 1.04]; 7-10 risk alleles: HR 0.95 [95% CI 0.90, 0.99]). None of these associations were statistically significant after correction for multiple testing. CONCLUSIONS/INTERPRETATION Our large-scale case-cohort study provides some evidence for a possible interaction of TCF7L2 variants and an incretin-specific genetic risk score with coffee consumption in relation to the risk of type 2 diabetes. Further large-scale studies and/or meta-analyses are needed to confirm these interactions in other populations.
Collapse
Affiliation(s)
- The InterAct Consortium
- c/o A. Heraclides, University of Nicosia Medical School, Centre for Primary Care and Population Health, 21 Ilia Papakyriakou, Engomi, P.O. Box 24005, 1700 Nicosia Cyprus
| |
Collapse
|
276
|
Kleinberger JW, Maloney KA, Pollin TI. The Genetic Architecture of Diabetes in Pregnancy: Implications for Clinical Practice. Am J Perinatol 2016; 33:1319-1326. [PMID: 27571483 PMCID: PMC5507691 DOI: 10.1055/s-0036-1592078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The genetic architecture of diabetes mellitus in general and in pregnancy is complex, owing to the multiple types of diabetes that comprise both complex/polygenic forms and monogenic (largely caused by a mutation in a single gene) forms such as maturity-onset diabetes of the young (MODY). Type 1 diabetes (T1D) and type 2 diabetes (T2D) have complex genetic etiologies, with over 40 and 90 genes/loci, respectively, implicated that interact with environmental/lifestyle factors. The genetic etiology of gestational diabetes mellitus has largely been found to overlap that of T2D. Genetic testing for complex forms of diabetes is not currently useful clinically, but genetic testing for monogenic forms, particularly MODY, has important utility for determining treatment, managing risk in family members, and pregnancy management. In particular, diagnosing MODY2, caused by GCK mutations, indicates that insulin should not be used, including during pregnancy, with the possible exception of an unaffected pregnancy during the third trimester to prevent macrosomia. A relatively simple method for identifying women with MODY2 has been piloted. MODY1, caused by HNF4A mutations, can paradoxically cause neonatal hyperinsulinemic hypoglycemia and macrosomia, indicating that detecting these cases is also clinically important. Diagnosing all MODY types provides opportunities for diagnosing other family members.
Collapse
Affiliation(s)
| | | | - Toni I. Pollin
- To Whom Correspondence May Be Addressed: Toni I. Pollin, MS, PhD, 660 West Redwood Street, Room 445C, Baltimore, MD 21201, 410-706-1630,
| |
Collapse
|
277
|
Villarroya F, Peyrou M, Giralt M. Transcriptional regulation of the uncoupling protein-1 gene. Biochimie 2016; 134:86-92. [PMID: 27693079 DOI: 10.1016/j.biochi.2016.09.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/25/2016] [Indexed: 02/08/2023]
Abstract
Regulated transcription of the uncoupling protein-1 (UCP1) gene, and subsequent UCP1 protein synthesis, is a hallmark of the acquisition of the differentiated, thermogenically competent status of brown and beige/brite adipocytes, as well as of the responsiveness of brown and beige/brite adipocytes to adaptive regulation of thermogenic activity. The 5' non-coding region of the UCP1 gene contains regulatory elements that confer tissue specificity, differentiation dependence, and neuro-hormonal regulation to UCP1 gene transcription. Two main regions-a distal enhancer and a proximal promoter region-mediate transcriptional regulation through interactions with a plethora of transcription factors, including nuclear hormone receptors and cAMP-responsive transcription factors. Co-regulators, such as PGC-1α, play a pivotal role in the concerted regulation of UCP1 gene transcription. Multiple interactions of transcription factors and co-regulators at the promoter region of the UCP1 gene result in local chromatin remodeling, leading to activation and increased accessibility of RNA polymerase II and subsequent gene transcription. Moreover, a commonly occurring A-to-G polymorphism in close proximity to the UCP1 gene enhancer influences the extent of UCP1 gene transcription. Notably, it has been reported that specific aspects of obesity and associated metabolic diseases are associated with human population variability at this site. On another front, the unique properties of the UCP1 promoter region have been exploited to develop brown adipose tissue-specific gene delivery tools for experimental purposes.
Collapse
Affiliation(s)
- Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain.
| | - Marion Peyrou
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Marta Giralt
- Department of Biochemistry and Molecular Biomedicine, Institut de Biomedicina (IBUB), University of Barcelona, Barcelona, Catalonia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain; Institut de Recerca Pediàtrica Sant Joan de Déu, Barcelona, Catalonia, Spain
| |
Collapse
|
278
|
Morita H, Komuro I. A Strategy for Genomic Research on Common Cardiovascular Diseases Aiming at the Realization of Precision Medicine. Circ Res 2016; 119:900-3. [DOI: 10.1161/circresaha.116.309802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Hiroyuki Morita
- From the Department of Cardiovascular Medicine, University of Tokyo, Japan
| | - Issei Komuro
- From the Department of Cardiovascular Medicine, University of Tokyo, Japan
| |
Collapse
|
279
|
Hasin-Brumshtein Y, Khan AH, Hormozdiari F, Pan C, Parks BW, Petyuk VA, Piehowski PD, Brümmer A, Pellegrini M, Xiao X, Eskin E, Smith RD, Lusis AJ, Smith DJ. Hypothalamic transcriptomes of 99 mouse strains reveal trans eQTL hotspots, splicing QTLs and novel non-coding genes. eLife 2016; 5. [PMID: 27623010 PMCID: PMC5053804 DOI: 10.7554/elife.15614] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
Previous studies had shown that the integration of genome wide expression profiles, in metabolic tissues, with genetic and phenotypic variance, provided valuable insight into the underlying molecular mechanisms. We used RNA-Seq to characterize hypothalamic transcriptome in 99 inbred strains of mice from the Hybrid Mouse Diversity Panel (HMDP), a reference resource population for cardiovascular and metabolic traits. We report numerous novel transcripts supported by proteomic analyses, as well as novel non coding RNAs. High resolution genetic mapping of transcript levels in HMDP, reveals both local and trans expression Quantitative Trait Loci (eQTLs) demonstrating 2 trans eQTL 'hotspots' associated with expression of hundreds of genes. We also report thousands of alternative splicing events regulated by genetic variants. Finally, comparison with about 150 metabolic and cardiovascular traits revealed many highly significant associations. Our data provide a rich resource for understanding the many physiologic functions mediated by the hypothalamus and their genetic regulation. DOI:http://dx.doi.org/10.7554/eLife.15614.001 Metabolism is a term that describes all the chemical reactions that are involved in keeping a living organism alive. Diseases related to metabolism – such as obesity, heart disease and diabetes – are a major health problem in the Western world. The causes of these diseases are complex and include both environmental factors, such as diet and exercise, and genetics. Indeed, many genetic variants that contribute to obesity have been uncovered in both humans and mice. However, it is only dimly understood how these genetic variants affect the underlying networks of interacting genes that cause metabolic disorders. Measuring gene activity or expression, and tracing how genetic instructions are carried from DNA into RNA and proteins, can reliably identify groups of genes that correlate with metabolic traits in specific organs. This strategy was successfully used in previous studies to reveal new information about abnormalities linked to obesity in specific tissues such as the liver and fat tissues. It was also shown that this approach might suggest new molecules that could be targeted to treat metabolic disorders. A brain region called the hypothalamus is key to the control of metabolism, including feeding behavior and obesity. Hasin-Brumshtein et al. set out to explore gene expression in the hypothalamus of 99 different strains of mice, in the hope that the data will help identify new connections between gene expression and metabolism. This approach showed that thousands of new and known genes are expressed in the mouse hypothalamus, some of which coded for proteins, and some of which did not. Hasin-Brumshtein et al. uncovered two genetic variants that controlled the expression of hundreds of other genes. Further analysis then revealed thousands of genetic variants that regulated the expression of, and type of RNA (so-called "spliceforms") produced from neighboring genes. Also, the expression of many individual genes showed significant similarities with about 150 metabolic measurements that had been evaluated previously in the mice. This new dataset is a unique resource that can be coupled with different approaches to test existing ideas and develop new ones about the role of particular genes or genetic mechanisms in obesity. Future studies will likely focus on new genes that show strong associations with attributes that are relevant to metabolic disorders, such as insulin levels, weight and fat mass. DOI:http://dx.doi.org/10.7554/eLife.15614.002
Collapse
Affiliation(s)
- Yehudit Hasin-Brumshtein
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, United States.,David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Microbiology, University of California, Los Angeles, Los Angeles, United states.,Department of Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Arshad H Khan
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Farhad Hormozdiari
- Department of Computer Science, University of California, Los Angeles, Los Angeles, United States
| | - Calvin Pan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, United States.,David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Brian W Parks
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, United States.,David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Microbiology, University of California, Los Angeles, Los Angeles, United states.,Department of Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, United States
| | - Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, United States
| | - Anneke Brümmer
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, United States
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, United States
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, United States
| | - Aldons J Lusis
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, United States.,David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Microbiology, University of California, Los Angeles, Los Angeles, United states.,Department of Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Desmond J Smith
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
280
|
Tsuneki H, Sasaoka T, Sakurai T. Sleep Control, GPCRs, and Glucose Metabolism. Trends Endocrinol Metab 2016; 27:633-642. [PMID: 27461005 DOI: 10.1016/j.tem.2016.06.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022]
Abstract
Modern lifestyles prolong daily activities into the nighttime, disrupting circadian rhythms, which may cause sleep disturbances. Sleep disturbances have been implicated in the dysregulation of blood glucose levels and reported to increase the risk of type 2 diabetes (T2D) and diabetic complications. Sleep disorders are treated using anti-insomnia drugs that target ionotropic and G protein-coupled receptors (GPCRs), including γ-aminobutyric acid (GABA) agonists, melatonin agonists, and orexin receptor antagonists. A deeper understanding of the effects of these medications on glucose metabolism and their underlying mechanisms of action is crucial for the treatment of diabetic patients with sleep disorders. In this review we focus on the beneficial impact of sleep on glucose metabolism and suggest a possible strategy for therapeutic intervention against sleep-related metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
281
|
Glastonbury C, Viñuela A, Buil A, Halldorsson G, Thorleifsson G, Helgason H, Thorsteinsdottir U, Stefansson K, Dermitzakis E, Spector T, Small K. Adiposity-Dependent Regulatory Effects on Multi-tissue Transcriptomes. Am J Hum Genet 2016; 99:567-579. [PMID: 27588447 PMCID: PMC5011064 DOI: 10.1016/j.ajhg.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 10/25/2022] Open
Abstract
Obesity is a global epidemic that is causally associated with a range of diseases, including type 2 diabetes and cardiovascular disease, at the population-level. However, there is marked heterogeneity in obesity-related outcomes among individuals. This might reflect genotype-dependent responses to adiposity. Given that adiposity, measured by BMI, is associated with widespread changes in gene expression and regulatory variants mediate the majority of known complex trait loci, we sought to identify gene-by-BMI (G × BMI) interactions on the regulation of gene expression in a multi-tissue RNA-sequencing (RNA-seq) dataset from the TwinsUK cohort (n = 856). At a false discovery rate of 5%, we identified 16 cis G × BMI interactions (top cis interaction: CHURC1, rs7143432, p = 2.0 × 10(-12)) and one variant regulating 53 genes in trans (top trans interaction: ZNF423, rs3851570, p = 8.2 × 10(-13)), all in adipose tissue. The interactions were adipose-specific and enriched for variants overlapping adipocyte enhancers, and regulated genes were enriched for metabolic and inflammatory processes. We replicated a subset of the interactions in an independent adipose RNA-seq dataset (deCODE genetics, n = 754). We also confirmed the interactions with an alternate measure of obesity, dual-energy X-ray absorptiometry (DXA)-derived visceral-fat-volume measurements, in a subset of TwinsUK individuals (n = 682). The identified G × BMI regulatory effects demonstrate the dynamic nature of gene regulation and reveal a functional mechanism underlying the heterogeneous response to obesity. Additionally, we have provided a web browser allowing interactive exploration of the dataset, including of association between expression, BMI, and G × BMI regulatory effects in four tissues.
Collapse
|
282
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
283
|
Abstract
The genetic architecture of common traits, including the number, frequency, and effect sizes of inherited variants that contribute to individual risk, has been long debated. Genome-wide association studies have identified scores of common variants associated with type 2 diabetes, but in aggregate, these explain only a fraction of the heritability of this disease. Here, to test the hypothesis that lower-frequency variants explain much of the remainder, the GoT2D and T2D-GENES consortia performed whole-genome sequencing in 2,657 European individuals with and without diabetes, and exome sequencing in 12,940 individuals from five ancestry groups. To increase statistical power, we expanded the sample size via genotyping and imputation in a further 111,548 subjects. Variants associated with type 2 diabetes after sequencing were overwhelmingly common and most fell within regions previously identified by genome-wide association studies. Comprehensive enumeration of sequence variation is necessary to identify functional alleles that provide important clues to disease pathophysiology, but large-scale sequencing does not support the idea that lower-frequency variants have a major role in predisposition to type 2 diabetes.
Collapse
|
284
|
Mehta ZB, Fine N, Pullen TJ, Cane MC, Hu M, Chabosseau P, Meur G, Velayos-Baeza A, Monaco AP, Marselli L, Marchetti P, Rutter GA. Changes in the expression of the type 2 diabetes-associated gene VPS13C in the β-cell are associated with glucose intolerance in humans and mice. Am J Physiol Endocrinol Metab 2016; 311:E488-507. [PMID: 27329800 PMCID: PMC5005967 DOI: 10.1152/ajpendo.00074.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
Abstract
Single nucleotide polymorphisms (SNPs) close to the VPS13C, C2CD4A and C2CD4B genes on chromosome 15q are associated with impaired fasting glucose and increased risk of type 2 diabetes. eQTL analysis revealed an association between possession of risk (C) alleles at a previously implicated causal SNP, rs7163757, and lowered VPS13C and C2CD4A levels in islets from female (n = 40, P < 0.041) but not from male subjects. Explored using promoter-reporter assays in β-cells and other cell lines, the risk variant at rs7163757 lowered enhancer activity. Mice deleted for Vps13c selectively in the β-cell were generated by crossing animals bearing a floxed allele at exon 1 to mice expressing Cre recombinase under Ins1 promoter control (Ins1Cre). Whereas Vps13c(fl/fl):Ins1Cre (βVps13cKO) mice displayed normal weight gain compared with control littermates, deletion of Vps13c had little effect on glucose tolerance. Pancreatic histology revealed no significant change in β-cell mass in KO mice vs. controls, and glucose-stimulated insulin secretion from isolated islets was not altered in vitro between control and βVps13cKO mice. However, a tendency was observed in female null mice for lower insulin levels and β-cell function (HOMA-B) in vivo. Furthermore, glucose-stimulated increases in intracellular free Ca(2+) were significantly increased in islets from female KO mice, suggesting impaired Ca(2+) sensitivity of the secretory machinery. The present data thus provide evidence for a limited role for changes in VPS13C expression in conferring altered disease risk at this locus, particularly in females, and suggest that C2CD4A may also be involved.
Collapse
Affiliation(s)
- Zenobia B Mehta
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Nicholas Fine
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Timothy J Pullen
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Matthew C Cane
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Pauline Chabosseau
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Gargi Meur
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | | | - Anthony P Monaco
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom; and
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom;
| |
Collapse
|
285
|
Morris DL, Sheng Y, Zhang Y, Wang YF, Zhu Z, Tombleson P, Chen L, Cunninghame Graham DS, Bentham J, Roberts AL, Chen R, Zuo X, Wang T, Wen L, Yang C, Liu L, Yang L, Li F, Huang Y, Yin X, Yang S, Rönnblom L, Fürnrohr BG, Voll RE, Schett G, Costedoat-Chalumeau N, Gaffney PM, Lau YL, Zhang X, Yang W, Cui Y, Vyse TJ. Genome-wide association meta-analysis in Chinese and European individuals identifies ten new loci associated with systemic lupus erythematosus. Nat Genet 2016; 48:940-946. [PMID: 27399966 PMCID: PMC4966635 DOI: 10.1038/ng.3603] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE; OMIM 152700) is a genetically complex autoimmune disease. Genome-wide association studies (GWASs) have identified more than 50 loci as robustly associated with the disease in single ancestries, but genome-wide transancestral studies have not been conducted. We combined three GWAS data sets from Chinese (1,659 cases and 3,398 controls) and European (4,036 cases and 6,959 controls) populations. A meta-analysis of these studies showed that over half of the published SLE genetic associations are present in both populations. A replication study in Chinese (3,043 cases and 5,074 controls) and European (2,643 cases and 9,032 controls) subjects found ten previously unreported SLE loci. Our study provides further evidence that the majority of genetic risk polymorphisms for SLE are contained within the same regions across both populations. Furthermore, a comparison of risk allele frequencies and genetic risk scores suggested that the increased prevalence of SLE in non-Europeans (including Asians) has a genetic basis.
Collapse
Affiliation(s)
- David L Morris
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Yujun Sheng
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Yan Zhang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Zhengwei Zhu
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Philip Tombleson
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Lingyan Chen
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | | | - James Bentham
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Amy L Roberts
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Ruoyan Chen
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xianbo Zuo
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Tingyou Wang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Leilei Wen
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Chao Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lu Liu
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lulu Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Feng Li
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Yuanbo Huang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Xianyong Yin
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Sen Yang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lars Rönnblom
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Barbara G Fürnrohr
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
- Division of Genetic Epidemiology, Medical University Innsbruck, Innsbruck, Austria
- Division of Biological Chemistry, Medical University Innsbruck, Innsbruck, Austria
| | - Reinhard E Voll
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
- Department of Rheumatology, University Hospital Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, University Hospital Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency, University Hospital Freiburg, Freiburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
- Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nathalie Costedoat-Chalumeau
- AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, Paris, France
- Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Xuejun Zhang
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yong Cui
- Department of Dermatology, No. 1 Hospital, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Timothy J Vyse
- Division of Genetics and Molecular Medicine, King's College London, London, UK
- Division of Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| |
Collapse
|
286
|
Simon PHG, Sylvestre MP, Tremblay J, Hamet P. Key Considerations and Methods in the Study of Gene-Environment Interactions. Am J Hypertens 2016; 29:891-9. [PMID: 27037711 DOI: 10.1093/ajh/hpw021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/08/2016] [Indexed: 12/16/2022] Open
Abstract
With increased involvement of genetic data in most epidemiological investigations, gene-environment (G × E) interactions now stand as a topic, which must be meticulously assessed and thoroughly understood. The level, mode, and outcomes of interactions between environmental factors and genetic traits have the capacity to modulate disease risk. These must, therefore, be carefully evaluated as they have the potential to offer novel insights on the "missing heritability problem", reaching beyond our current limitations. First, we review a definition of G × E interactions. We then explore how concepts such as the early manifestation of the genetic components of a disease, the heterogeneity of complex traits, the clear definition of epidemiological strata, and the effect of varying physiological conditions can affect our capacity to detect (or miss) G × E interactions. Lastly, we discuss the shortfalls of regression models to study G × E interactions and how other methods such as the ReliefF algorithm, pattern recognition methods, or the LASSO (Least Absolute Shrinkage and Selection Operator) method can enable us to more adequately model G × E interactions. Overall, we present the elements to consider and a path to follow when studying genetic determinants of disease in order to uncover potential G × E interactions.
Collapse
Affiliation(s)
- Paul H G Simon
- CHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Marie-Pierre Sylvestre
- CHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Johanne Tremblay
- CHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Pavel Hamet
- CHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
287
|
Kahraman S, Okawa ER, Kulkarni RN. Is Transforming Stem Cells to Pancreatic Beta Cells Still the Holy Grail for Type 2 Diabetes? Curr Diab Rep 2016; 16:70. [PMID: 27313072 PMCID: PMC5877461 DOI: 10.1007/s11892-016-0764-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes is a progressive disease affecting millions of people worldwide. There are several medications and treatment options to improve the life quality of people with diabetes. One of the strategies for the treatment of diabetes could be the use of human pluripotent stem cells or induced pluripotent stem cells. The recent advances in differentiation of stem cells into insulin-secreting beta-like cells in vitro make the transplantation of the stem cell-derived beta-like cells an attractive approach for treatment of type 1 and type 2 diabetes. While stem cell-derived beta-like cells provide an unlimited cell source for beta cell replacement therapies, these cells can also be used as a platform for drug screening or modeling diseases.
Collapse
Affiliation(s)
- Sevim Kahraman
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Erin R Okawa
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Boston, MA, 02215, USA
| | - Rohit N Kulkarni
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA.
- Harvard Stem Cell Institute, Boston, MA, 02215, USA.
| |
Collapse
|
288
|
Beer NL, Gloyn AL. Genome-edited human stem cell-derived beta cells: a powerful tool for drilling down on type 2 diabetes GWAS biology. F1000Res 2016; 5:F1000 Faculty Rev-1711. [PMID: 27508066 PMCID: PMC4955023 DOI: 10.12688/f1000research.8682.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes (T2D) is a disease of pandemic proportions, one defined by a complex aetiological mix of genetic, epigenetic, environmental, and lifestyle risk factors. Whilst the last decade of T2D genetic research has identified more than 100 loci showing strong statistical association with disease susceptibility, our inability to capitalise upon these signals reflects, in part, a lack of appropriate human cell models for study. This review discusses the impact of two complementary, state-of-the-art technologies on T2D genetic research: the generation of stem cell-derived, endocrine pancreas-lineage cells and the editing of their genomes. Such models facilitate investigation of diabetes-associated genomic perturbations in a physiologically representative cell context and allow the role of both developmental and adult islet dysfunction in T2D pathogenesis to be investigated. Accordingly, we interrogate the role that patient-derived induced pluripotent stem cell models are playing in understanding cellular dysfunction in monogenic diabetes, and how site-specific nucleases such as the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system are helping to confirm genes crucial to human endocrine pancreas development. We also highlight the novel biology gleaned in the absence of patient lines, including an ability to model the whole phenotypic spectrum of diabetes phenotypes occurring both in utero and in adult cells, interrogating the non-coding 'islet regulome' for disease-causing perturbations, and understanding the role of other islet cell types in aberrant glycaemia. This article aims to reinforce the importance of investigating T2D signals in cell models reflecting appropriate species, genomic context, developmental time point, and tissue type.
Collapse
Affiliation(s)
- Nicola L. Beer
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,
| | - Anna L. Gloyn
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,Wellcome Trust Centre for Human Genetics, Oxford, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
289
|
Abstract
As with other complex diseases, unbiased association studies followed by physiological and experimental characterization have for years formed a paradigm for identifying genes or processes of relevance to type 2 diabetes mellitus (T2D). Recent large-scale common and rare variant genome-wide association studies (GWAS) suggest that substantially larger association studies are needed to identify most T2D loci in the population. To hasten clinical translation of genetic discoveries, new paradigms are also required to aid specialized investigation of nascent hypotheses. We argue for an integrated T2D knowledgebase, designed for a worldwide community to access aggregated large-scale genetic data sets, as one paradigm to catalyse convergence of these efforts.
Collapse
|
290
|
Thomas PPM, Alshehri SM, van Kranen HJ, Ambrosino E. The impact of personalized medicine of Type 2 diabetes mellitus in the global health context. Per Med 2016; 13:381-393. [DOI: 10.2217/pme-2016-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advances in the fields of genomic sciences have given rise to personalized medicine. This new paradigm draws upon a patient's genetic and metabolic makeup in order to tailor diagnostics and treatment. Personalized medicine holds remarkable promises to improve prevention and management of chronic diseases of global relevance, such as Type 2 diabetes mellitus (T2DM). This review article aims at summarizing the evidence from genome-based sciences on T2DM risk and management in different populations and in the Global Health context. Opinions from leading experts in the field were also included. Based on these findings, strengths and weaknesses of personalized approach to T2DM in a global context are delineated. Implications for future research and implementation on that subject are discussed.
Collapse
Affiliation(s)
- Pierre Paul Michel Thomas
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Salih Mohammed Alshehri
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Henk J van Kranen
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
- National Institute for Public Health & the Environment, Bilthoven 3721 MA, The Netherlands
| | - Elena Ambrosino
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| |
Collapse
|
291
|
Lin J, Hu Y, Nunez S, Foulkes AS, Cieply B, Xue C, Gerelus M, Li W, Zhang H, Rader DJ, Musunuru K, Li M, Reilly MP. Transcriptome-Wide Analysis Reveals Modulation of Human Macrophage Inflammatory Phenotype Through Alternative Splicing. Arterioscler Thromb Vasc Biol 2016; 36:1434-47. [PMID: 27230130 PMCID: PMC4919157 DOI: 10.1161/atvbaha.116.307573] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Human macrophages can shift phenotype across the inflammatory M1 and reparative M2 spectrum in response to environmental challenges, but the mechanisms promoting inflammatory and cardiometabolic disease-associated M1 phenotypes remain incompletely understood. Alternative splicing (AS) is emerging as an important regulator of cellular function, yet its role in macrophage activation is largely unknown. We investigated the extent to which AS occurs in M1 activation within the cardiometabolic disease context and validated a functional genomic cell model for studying human macrophage-related AS events. APPROACH AND RESULTS From deep RNA-sequencing of resting, M1, and M2 primary human monocyte-derived macrophages, we found 3860 differentially expressed genes in M1 activation and detected 233 M1-induced AS events; the majority of AS events were cell- and M1-specific with enrichment for pathways relevant to macrophage inflammation. Using genetic variant data for 10 cardiometabolic traits, we identified 28 trait-associated variants within the genomic loci of 21 alternatively spliced genes and 15 variants within 7 differentially expressed regulatory splicing factors in M1 activation. Knockdown of 1 such splicing factor, CELF1, in primary human macrophages led to increased inflammatory response to M1 stimulation, demonstrating CELF1's potential modulation of the M1 phenotype. Finally, we demonstrated that an induced pluripotent stem cell-derived macrophage system recapitulates M1-associated AS events and provides a high-fidelity macrophage AS model. CONCLUSIONS AS plays a role in defining macrophage phenotype in a cell- and stimulus-specific fashion. Alternatively spliced genes and splicing factors with trait-associated variants may reveal novel pathways and targets in cardiometabolic diseases.
Collapse
Affiliation(s)
- Jennie Lin
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.).
| | - Yu Hu
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Sara Nunez
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Andrea S Foulkes
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Benjamin Cieply
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Chenyi Xue
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Mark Gerelus
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Wenjun Li
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Hanrui Zhang
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Daniel J Rader
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Kiran Musunuru
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Mingyao Li
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.)
| | - Muredach P Reilly
- From the Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine (J.L.), Department of Biostatistics and Epidemiology (Y.H., M.L.), Department of Genetics, Perelman School of Medicine (B.C., K.M., D.J.R.), and Cardiovascular Institute, Department of Medicine, Perelman School of Medicine (M.G., W.L., K.M.), University of Pennsylvania, Philadelphia; Irving Institute for Clinical and Translational Research (M.P.R.) and Division of Cardiology, Department of Medicine (C.X., H.Z., M.P.R.), Columbia University Medical Center, New York, NY; and Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA (S.N., A.S.F.).
| |
Collapse
|
292
|
Mitchell JS, Li N, Weinhold N, Försti A, Ali M, van Duin M, Thorleifsson G, Johnson DC, Chen B, Halvarsson BM, Gudbjartsson DF, Kuiper R, Stephens OW, Bertsch U, Broderick P, Campo C, Einsele H, Gregory WA, Gullberg U, Henrion M, Hillengass J, Hoffmann P, Jackson GH, Johnsson E, Jöud M, Kristinsson SY, Lenhoff S, Lenive O, Mellqvist UH, Migliorini G, Nahi H, Nelander S, Nickel J, Nöthen MM, Rafnar T, Ross FM, da Silva Filho MI, Swaminathan B, Thomsen H, Turesson I, Vangsted A, Vogel U, Waage A, Walker BA, Wihlborg AK, Broyl A, Davies FE, Thorsteinsdottir U, Langer C, Hansson M, Kaiser M, Sonneveld P, Stefansson K, Morgan GJ, Goldschmidt H, Hemminki K, Nilsson B, Houlston RS. Genome-wide association study identifies multiple susceptibility loci for multiple myeloma. Nat Commun 2016; 7:12050. [PMID: 27363682 PMCID: PMC4932178 DOI: 10.1038/ncomms12050] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/24/2016] [Indexed: 02/08/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy with a significant heritable basis. Genome-wide association studies have transformed our understanding of MM predisposition, but individual studies have had limited power to discover risk loci. Here we perform a meta-analysis of these GWAS, add a new GWAS and perform replication analyses resulting in 9,866 cases and 239,188 controls. We confirm all nine known risk loci and discover eight new loci at 6p22.3 (rs34229995, P=1.31 × 10(-8)), 6q21 (rs9372120, P=9.09 × 10(-15)), 7q36.1 (rs7781265, P=9.71 × 10(-9)), 8q24.21 (rs1948915, P=4.20 × 10(-11)), 9p21.3 (rs2811710, P=1.72 × 10(-13)), 10p12.1 (rs2790457, P=1.77 × 10(-8)), 16q23.1 (rs7193541, P=5.00 × 10(-12)) and 20q13.13 (rs6066835, P=1.36 × 10(-13)), which localize in or near to JARID2, ATG5, SMARCD3, CCAT1, CDKN2A, WAC, RFWD3 and PREX1. These findings provide additional support for a polygenic model of MM and insight into the biological basis of tumour development.
Collapse
Affiliation(s)
- Jonathan S. Mitchell
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Ni Li
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Niels Weinhold
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
- Department of Internal Medicine V, University of Heidelberg, 69117 Heidelberg, Germany
| | - Asta Försti
- German Cancer Research Center, 69120 Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, SE-205 02 Malmo, Sweden
| | - Mina Ali
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Mark van Duin
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA Rotterdam, The Netherlands
| | | | - David C. Johnson
- Division of Molecular Pathology, The Institute of Cancer Research, Surrey SM2 5NG, UK
| | - Bowang Chen
- German Cancer Research Center, 69120 Heidelberg, Germany
| | - Britt-Marie Halvarsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Daniel F. Gudbjartsson
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, IS-101 Reykjavik, Iceland
| | - Rowan Kuiper
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA Rotterdam, The Netherlands
| | - Owen W. Stephens
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | - Uta Bertsch
- Department of Internal Medicine V, University of Heidelberg, 69117 Heidelberg, Germany
- National Centre of Tumor Diseases, 69120 Heidelberg, Germany
| | - Peter Broderick
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Chiara Campo
- German Cancer Research Center, 69120 Heidelberg, Germany
| | | | - Walter A. Gregory
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9PH, UK
| | - Urban Gullberg
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Marc Henrion
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Jens Hillengass
- Department of Internal Medicine V, University of Heidelberg, 69117 Heidelberg, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, D-53127 Bonn, Germany
- Division of Medical Genetics, Department of Biomedicine, University of Basel, 4003 Basel, Switzerland
| | | | - Ellinor Johnsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Magnus Jöud
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
- Clinical Immunology and Transfusion Medicine, Laboratory Medicine, Office of Medical Services, SE-221 85 Lund, Sweden
| | - Sigurður Y. Kristinsson
- Department of Hematology, Landspitali, National University Hospital of Iceland, IS-101 Reykjavik, Iceland
| | - Stig Lenhoff
- Hematology Clinic, Skåne University Hospital, SE-221 85 Lund, Sweden
| | - Oleg Lenive
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Ulf-Henrik Mellqvist
- Section of Hematology, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
| | - Gabriele Migliorini
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Hareth Nahi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Sven Nelander
- Rudbeck Laboratory, Department of Immunology, Pathology and Genetics, Uppsala University, SE-751 05 Uppsala, Sweden
| | - Jolanta Nickel
- Department of Internal Medicine V, University of Heidelberg, 69117 Heidelberg, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, D-53127 Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, D-53127 Bonn, Germany
| | - Thorunn Rafnar
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
| | - Fiona M. Ross
- Wessex Regional Genetics Laboratory, University of Southampton, Salisbury SP2 8BJ, UK
| | | | - Bhairavi Swaminathan
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Hauke Thomsen
- German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ingemar Turesson
- Hematology Clinic, Skåne University Hospital, SE-221 85 Lund, Sweden
| | - Annette Vangsted
- Department of Haematology, University Hospital of Copenhagen at Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Box 8905, N-7491 Trondheim, Norway
| | - Brian A. Walker
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | - Anna-Karin Wihlborg
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
| | - Annemiek Broyl
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA Rotterdam, The Netherlands
| | - Faith E. Davies
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, IS-101 Reykjavik, Iceland
| | - Christian Langer
- Department of Internal Medicine III, University of Ulm, D-89081 Ulm, Germany
| | - Markus Hansson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
- Hematology Clinic, Skåne University Hospital, SE-221 85 Lund, Sweden
| | - Martin Kaiser
- Division of Molecular Pathology, The Institute of Cancer Research, Surrey SM2 5NG, UK
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA Rotterdam, The Netherlands
| | | | - Gareth J. Morgan
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, 69117 Heidelberg, Germany
- National Centre of Tumor Diseases, 69120 Heidelberg, Germany
| | - Kari Hemminki
- German Cancer Research Center, 69120 Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, SE-205 02 Malmo, Sweden
| | - Björn Nilsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84 Lund, Sweden
- Clinical Immunology and Transfusion Medicine, Laboratory Medicine, Office of Medical Services, SE-221 85 Lund, Sweden
- Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| |
Collapse
|
293
|
Affiliation(s)
- R. Mrowka
- Experimentelle Nephrologie; Universitätsklinikum Jena, KIM III; Jena Germany
| | - S. Reuter
- Experimentelle Nephrologie; Universitätsklinikum Jena, KIM III; Jena Germany
| |
Collapse
|
294
|
Flannick J, Johansson S, Njølstad PR. Common and rare forms of diabetes mellitus: towards a continuum of diabetes subtypes. Nat Rev Endocrinol 2016; 12:394-406. [PMID: 27080136 DOI: 10.1038/nrendo.2016.50] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Insights into the genetic basis of type 2 diabetes mellitus (T2DM) have been difficult to discern, despite substantial research. More is known about rare forms of diabetes mellitus, several of which share clinical and genetic features with the common form of T2DM. In this Review, we discuss the extent to which the study of rare and low-frequency mutations in large populations has begun to bridge the gap between rare and common forms of diabetes mellitus. We hypothesize that the perceived division between these diseases might be due, in part, to the historical ascertainment bias of genetic studies, rather than a clear distinction between disease pathophysiologies. We also discuss possible implications of a new model for the genetic basis of diabetes mellitus subtypes, where the boundary between subtypes becomes blurred.
Collapse
Affiliation(s)
- Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Center for Human Genetic Research, Massachusetts General Hospital, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - Stefan Johansson
- K.G. Jebsen Center for Diabetes Research, The Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5020 Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Jonas Lies veg 65, N-5021 Bergen, Norway
| | - Pål R Njølstad
- K.G. Jebsen Center for Diabetes Research, The Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5020 Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Jonas Lies veg 65, N-5021 Bergen, Norway
| |
Collapse
|
295
|
Abstract
Large sample sizes, high-resolution arrays and comprehensive imputation are pushing genetic fine-mapping of complex trait loci to its limits without, in most cases, pinpointing a unique variant-gene combination. Superimposing these results on sophisticated maps of functional chromatin elements promises to break this logjam, as a new study of type 2 diabetes compellingly demonstrates.
Collapse
Affiliation(s)
- Constantin Polychronakos
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Maha Alriyami
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
296
|
Increased Melatonin Signaling Is a Risk Factor for Type 2 Diabetes. Cell Metab 2016; 23:1067-1077. [PMID: 27185156 DOI: 10.1016/j.cmet.2016.04.009] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/18/2016] [Accepted: 04/13/2016] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes (T2D) is a global pandemic. Genome-wide association studies (GWASs) have identified >100 genetic variants associated with the disease, including a common variant in the melatonin receptor 1 b gene (MTNR1B). Here, we demonstrate increased MTNR1B expression in human islets from risk G-allele carriers, which likely leads to a reduction in insulin release, increasing T2D risk. Accordingly, in insulin-secreting cells, melatonin reduced cAMP levels, and MTNR1B overexpression exaggerated the inhibition of insulin release exerted by melatonin. Conversely, mice with a disruption of the receptor secreted more insulin. Melatonin treatment in a human recall-by-genotype study reduced insulin secretion and raised glucose levels more extensively in risk G-allele carriers. Thus, our data support a model where enhanced melatonin signaling in islets reduces insulin secretion, leading to hyperglycemia and greater future risk of T2D. The findings also imply that melatonin physiologically serves to inhibit nocturnal insulin release.
Collapse
|
297
|
Lane JM, Chang AM, Bjonnes AC, Aeschbach D, Anderson C, Cade BE, Cain SW, Czeisler CA, Gharib SA, Gooley JJ, Gottlieb DJ, Grant SFA, Klerman EB, Lauderdale DS, Lockley SW, Munch M, Patel S, Punjabi NM, Rajaratnam SMW, Rueger M, St Hilaire MA, Santhi N, Scheuermaier K, Van Reen E, Zee PC, Shea SA, Duffy JF, Buxton OM, Redline S, Scheer FAJL, Saxena R. Impact of Common Diabetes Risk Variant in MTNR1B on Sleep, Circadian, and Melatonin Physiology. Diabetes 2016; 65:1741-51. [PMID: 26868293 PMCID: PMC4878414 DOI: 10.2337/db15-0999] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/07/2016] [Indexed: 12/23/2022]
Abstract
The risk of type 2 diabetes (T2D) is increased by abnormalities in sleep quantity and quality, circadian alignment, and melatonin regulation. A common genetic variant in a receptor for the circadian-regulated hormone melatonin (MTNR1B) is associated with increased fasting blood glucose and risk of T2D, but whether sleep or circadian disruption mediates this risk is unknown. We aimed to test if MTNR1B diabetes risk variant rs10830963 associates with measures of sleep or circadian physiology in intensive in-laboratory protocols (n = 58-96) or cross-sectional studies with sleep quantity and quality and timing measures from self-report (n = 4,307-10,332), actigraphy (n = 1,513), or polysomnography (n = 3,021). In the in-laboratory studies, we found a significant association with a substantially longer duration of elevated melatonin levels (41 min) and delayed circadian phase of dim-light melatonin offset (1.37 h), partially mediated through delayed offset of melatonin synthesis. Furthermore, increased T2D risk in MTNR1B risk allele carriers was more pronounced in early risers versus late risers as determined by 7 days of actigraphy. Our results provide the surprising insight that the MTNR1B risk allele influences dynamics of melatonin secretion, generating a novel hypothesis that the MTNR1B risk allele may extend the duration of endogenous melatonin production later into the morning and that early waking may magnify the diabetes risk conferred by the risk allele.
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| | - Anne-Marie Chang
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Department of Biobehavioral Health, Pennsylvania State University, University Park, PA
| | - Andrew C Bjonnes
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| | - Daniel Aeschbach
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Clare Anderson
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sean W Cain
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA
| | - Joshua J Gooley
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Struan F A Grant
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | | | - Steven W Lockley
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Miriam Munch
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Sanjay Patel
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Naresh M Punjabi
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Shanthakumar M W Rajaratnam
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Melanie Rueger
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Melissa A St Hilaire
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Nayantara Santhi
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Karin Scheuermaier
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Eliza Van Reen
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Phyllis C Zee
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Steven A Shea
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Orfeu M Buxton
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Department of Biobehavioral Health, Pennsylvania State University, University Park, PA Department of Social and Behavioral Sciences, Harvard School of Public Health, Boston, MA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Richa Saxena
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA Program in Medical and Population Genetics, Broad Institute, Cambridge, MA Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
298
|
|
299
|
Jin T. Current Understanding on Role of the Wnt Signaling Pathway Effector TCF7L2 in Glucose Homeostasis. Endocr Rev 2016; 37:254-77. [PMID: 27159876 DOI: 10.1210/er.2015-1146] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The role of the Wnt signaling pathway in metabolic homeostasis has drawn our intensive attention, especially after the genome-wide association study discovery that certain polymorphisms of its key effector TCF7L2 are strongly associated with the susceptibility to type 2 diabetes. For a decade, great efforts have been made in determining the function of TCF7L2 in various metabolic organs, which have generated both considerable achievements and disputes. In this review, I will briefly introduce the canonical Wnt signaling pathway, focusing on its effector β-catenin/TCF, including emphasizing the bidirectional feature of TCFs and β-catenin post-translational modifications. I will then summarize the observations on the association between TCF7L2 polymorphisms and type 2 diabetes risk. The main content, however, is on the intensive functional exploration of the metabolic role of TCF7L2, including the disputes generated on determining its role in the pancreas and liver with various transgenic mouse lines. Finally, I will discuss those achievements and disputes and present my future perspectives.
Collapse
Affiliation(s)
- Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
300
|
Affiliation(s)
- Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|