151
|
Morris GE, Braund PS, Moore JS, Samani NJ, Codd V, Webb TR. Coronary Artery Disease-Associated LIPA Coding Variant rs1051338 Reduces Lysosomal Acid Lipase Levels and Activity in Lysosomes. Arterioscler Thromb Vasc Biol 2017; 37:1050-1057. [PMID: 28279971 PMCID: PMC5444428 DOI: 10.1161/atvbaha.116.308734] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Genome-wide association studies have linked variants at chromosome 10q23 with increased coronary artery disease risk. The disease-associated variants fall in LIPA, which encodes lysosomal acid lipase (LAL), the enzyme responsible for lysosomal cholesteryl ester hydrolysis. Loss-of-function mutations in LIPA result in accelerated atherosclerosis. Surprisingly, the coronary artery disease variants are associated with increased LIPA expression in some cell types. In this study, we address this apparent contradiction. Approach and Results— We investigated a coding variant rs1051338, which is in high linkage disequilibrium (r2=0.89) with the genome-wide association study lead–associated variant rs2246833 and causes a nonsynonymous threonine to proline change within the signal peptide of LAL. Transfection of allele-specific expression constructs showed that the risk allele results in reduced lysosomal LAL protein (P=0.004) and activity (P=0.005). Investigation of LAL localization and turnover showed the risk LAL protein is degraded more quickly. This mechanism was confirmed in disease-relevant macrophages from individuals homozygous for either the nonrisk or risk allele. There was no difference in LAL protein or activity in whole macrophage extracts; however, we found reduced LAL protein (P=0.02) and activity (P=0.026) with the risk genotype in lysosomal extracts, suggesting that the risk genotype affects lysosomal LAL activity. Inhibition of the proteasome resulted in equal amounts of lysosomal LAL protein in risk and nonrisk macrophages. Conclusions— Our findings show that the coronary artery disease–associated coding variant rs1051338 causes reduced lysosomal LAL protein and activity because of increased LAL degradation, providing a plausible causal mechanism of increased coronary artery disease risk.
Collapse
Affiliation(s)
- Gavin E Morris
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom
| | - Peter S Braund
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom
| | - Jasbir S Moore
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom
| | - Nilesh J Samani
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom
| | - Veryan Codd
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom
| | - Tom R Webb
- From the Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, United Kingdom.
| |
Collapse
|
152
|
Atzler D, Appelbaum S, Cordts K, Ojeda FM, Wild PS, Münzel T, Blankenberg S, Böger RH, Blettner M, Beutel ME, Pfeiffer N, Zeller T, Lackner KJ, Schwedhelm E. Reference intervals of plasma homoarginine from the German Gutenberg Health Study. Clin Chem Lab Med 2017; 54:1231-7. [PMID: 26562034 DOI: 10.1515/cclm-2015-0785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/12/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Low circulating homoarginine has been associated with adverse cardiovascular (CV) outcome and mortality in patients at risk and in the general population. The present study aimed to define plasma homoarginine reference intervals from a representative population sample to improve risk stratification between healthy individuals and individuals at risk. METHODS We determined age- and sex-specific reference intervals for circulating plasma homoarginine in a subgroup of 786 healthy participants (no CV disease or risk factors) of the Gutenberg Health Study. Homoarginine concentrations were measured using a validated liquid chromatography-tandem mass spectrometry method. RESULTS Median EDTA plasma homoarginine concentration was 1.88 [25th; 75th percentile, 1.47; 2.41] μmol/L, with lower concentrations in women (1.77 [1.38; 2.26] μmol/L) than in men (2.01 [1.61; 2.56] μmol/L; p<0.001). Sex-specific 2.5th and 97.5th percentiles of reference intervals were 0.84 and 3.89 μmol/L in women and 0.98 and 4.10 μmol/L in men, respectively. Homoarginine concentrations also depended on age and single nucleotide polymorphisms related to the L-arginine:glycine amidinotransferase gene. CONCLUSIONS We provide plasma homoarginine reference intervals in men and women of the general population. The determination of homoarginine levels might be favorable for individual risk stratification.
Collapse
|
153
|
Hoffman JD, Graff RE, Emami NC, Tai CG, Passarelli MN, Hu D, Huntsman S, Hadley D, Leong L, Majumdar A, Zaitlen N, Ziv E, Witte JS. Cis-eQTL-based trans-ethnic meta-analysis reveals novel genes associated with breast cancer risk. PLoS Genet 2017; 13:e1006690. [PMID: 28362817 PMCID: PMC5391966 DOI: 10.1371/journal.pgen.1006690] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/14/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common solid organ malignancy and the most frequent cause of cancer death among women worldwide. Previous research has yielded insights into its genetic etiology, but there remains a gap in the understanding of genetic factors that contribute to risk, and particularly in the biological mechanisms by which genetic variation modulates risk. The National Cancer Institute's "Up for a Challenge" (U4C) competition provided an opportunity to further elucidate the genetic basis of the disease. Our group leveraged the seven datasets made available by the U4C organizers and data from the publicly available UK Biobank cohort to examine associations between imputed gene expression and breast cancer risk. In particular, we used reference datasets describing the breast tissue and whole blood transcriptomes to impute expression levels in breast cancer cases and controls. In trans-ethnic meta-analyses of U4C and UK Biobank data, we found significant associations between breast cancer risk and the expression of RCCD1 (joint p-value: 3.6x10-06) and DHODH (p-value: 7.1x10-06) in breast tissue, as well as a suggestive association for ANKLE1 (p-value: 9.3x10-05). Expression of RCCD1 in whole blood was also suggestively associated with disease risk (p-value: 1.2x10-05), as were expression of ACAP1 (p-value: 1.9x10-05) and LRRC25 (p-value: 5.2x10-05). While genome-wide association studies (GWAS) have implicated RCCD1 and ANKLE1 in breast cancer risk, they have not identified the remaining three genes. Among the genetic variants that contributed to the predicted expression of the five genes, we found 23 nominally (p-value < 0.05) associated with breast cancer risk, among which 15 are not in high linkage disequilibrium with risk variants previously identified by GWAS. In summary, we used a transcriptome-based approach to investigate the genetic underpinnings of breast carcinogenesis. This approach provided an avenue for deciphering the functional relevance of genes and genetic variants involved in breast cancer.
Collapse
Affiliation(s)
- Joshua D. Hoffman
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Rebecca E. Graff
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Nima C. Emami
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
- Program in Biological and Medical Informatics, University of California San Francisco, San Francisco, CA, United States of America
| | - Caroline G. Tai
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Michael N. Passarelli
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States of America
| | - Donglei Hu
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Scott Huntsman
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Dexter Hadley
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States of America
| | - Lancelote Leong
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Arunabha Majumdar
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - Noah Zaitlen
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Elad Ziv
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States of America
- Department of Urology, University of California San Francisco, San Francisco, CA, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
154
|
Fava VM, Sales-Marques C, Alcaïs A, Moraes MO, Schurr E. Age-Dependent Association of TNFSF15/ TNFSF8 Variants and Leprosy Type 1 Reaction. Front Immunol 2017; 8:155. [PMID: 28261213 PMCID: PMC5306391 DOI: 10.3389/fimmu.2017.00155] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/30/2017] [Indexed: 11/21/2022] Open
Abstract
A current major challenge in leprosy control is the prevention of permanent disabilities. Host pathological inflammatory responses termed type 1 reaction (T1R) are a leading cause of nerve damage for leprosy patients. The environmental or inherited factors that predispose leprosy cases to undergo T1R are not known. However, studies have shown an important contribution of host genetics for susceptibility to T1R. We have previously identified variants encompassing the TNFSF15/TNFSF8 genes as T1R risk factors in a Vietnamese sample and replicated this association in a Brazilian sample. However, we failed to validate in Brazilian patients the strong association of TNFSF15/TNFSF8 markers rs6478108 and rs7863183 with T1R that we had observed in Vietnamese patients. Here, we investigated if the lack of validation of these variants was due to age-dependent effects on association using four independent population samples, two from Brazil and two from Vietnam. In the combined analysis across the four samples, we observed a strong association of the TNFSF15/TNFSF8 variants rs6478108, rs7863183, and rs3181348 with T1R (pcombined = 1.5E−05, pcombined = 1.8E−05, and pcombined = 6.5E−06, respectively). However, the association of rs6478108 with T1R was more pronounced in leprosy cases under 30 years of age compared to the global sample [odds ratio (OR) = 1.95, 95% confidence interval (CI) = 1.54–2.46, pcombined = 2.5E−08 versus OR = 1.46, 95% CI = 1.23–1.73, pcombined = 1.5E−05]. A multivariable analysis indicated that the association of rs6478108 with T1R was independent of either rs7863183 or rs3181348. These three variants are known regulators of the TNFSF8 gene transcription level in multiple tissues. The age dependency of association of rs6478108 and T1R suggests that the genetic control of gene expression varies across the human life span.
Collapse
Affiliation(s)
- Vinicius M Fava
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; The McGill International TB Centre, Department of Human Genetics, McGill University, Montreal, QC, Canada
| | | | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France; Imagine Institute, University Paris Descartes, Paris, France; Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Milton O Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ , Rio de Janeiro , Brazil
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; The McGill International TB Centre, Department of Human Genetics, McGill University, Montreal, QC, Canada; Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
155
|
Ovenden ES, Drögemöller BI, van der Merwe L, Chiliza B, Asmal L, Emsley RA, Warnich L. Fine-mapping of antipsychotic response genome-wide association studies reveals novel regulatory mechanisms. Pharmacogenomics 2017; 18:105-120. [DOI: 10.2217/pgs-2016-0108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aim: Noncoding variation has demonstrated regulatory effects on disease treatment outcomes. This study investigated the potential functionality of previously implicated noncoding variants on schizophrenia treatment response. Materials & methods: Predicted regulatory potential of variation identified from antipsychotic response genome-wide association studies was determined. Prioritized variants were assessed for association(s) with treatment outcomes in a South African first episode schizophrenia cohort (n = 103). Results: Bioinformatic and association results implicated a relationship between regulatory variants, expression of MANBA, COL9A2 and NFKB1, and treatment response. Three SNPs were associated with poor outcomes (rs230493: p = 1.88 × 10-6; rs3774959: p = 1.75 × 10-5; and rs230504: p = 1.48 × 10-4). Conclusion: This study has thoroughly investigated previous GWAS to pinpoint variants that may play a causal role in poor schizophrenia treatment outcomes, and provides potential candidate genes for further study in the field of antipsychotic response.
Collapse
Affiliation(s)
- Ellen S Ovenden
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | | | - Lize van der Merwe
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Bonginkosi Chiliza
- Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Laila Asmal
- Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Robin A Emsley
- Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Louise Warnich
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
156
|
Choudhury M, Ramsey SA. Identifying Cell Type-Specific Transcription Factors by Integrating ChIP-seq and eQTL Data-Application to Monocyte Gene Regulation. GENE REGULATION AND SYSTEMS BIOLOGY 2016; 10:105-110. [PMID: 28008225 PMCID: PMC5156548 DOI: 10.4137/grsb.s40768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/03/2016] [Accepted: 11/06/2016] [Indexed: 01/22/2023]
Abstract
We describe a novel computational approach to identify transcription factors (TFs) that are candidate regulators in a human cell type of interest. Our approach involves integrating cell type-specific expression quantitative trait locus (eQTL) data and TF data from chromatin immunoprecipitation-to-tag-sequencing (ChIP-seq) experiments in cell lines. To test the method, we used eQTL data from human monocytes in order to screen for TFs. Using a list of known monocyte-regulating TFs, we tested the hypothesis that the binding sites of cell type-specific TF regulators would be concentrated in the vicinity of monocyte eQTLs. For each of 397 ChIP-seq data sets, we obtained an enrichment ratio for the number of ChIP-seq peaks that are located within monocyte eQTLs. We ranked ChIP-seq data sets according to their statistical significances for eQTL overlap, and from this ranking, we observed that monocyte-regulating TFs are more highly ranked than would be expected by chance. We identified 27 TFs that had significant monocyte enrichment scores and mapped them into a protein interaction network. Our analysis uncovered two novel candidate monocyte-regulating TFs, BCLAF1 and SIN3A. Our approach is an efficient method to identify candidate TFs that can be used for any cell/tissue type for which eQTL data are available.
Collapse
Affiliation(s)
- Mudra Choudhury
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
157
|
Ried JS, Jeff M. J, Chu AY, Bragg-Gresham JL, van Dongen J, Huffman JE, Ahluwalia TS, Cadby G, Eklund N, Eriksson J, Esko T, Feitosa MF, Goel A, Gorski M, Hayward C, Heard-Costa NL, Jackson AU, Jokinen E, Kanoni S, Kristiansson K, Kutalik Z, Lahti J, Luan J, Mägi R, Mahajan A, Mangino M, Medina-Gomez C, Monda KL, Nolte IM, Pérusse L, Prokopenko I, Qi L, Rose LM, Salvi E, Smith MT, Snieder H, Stančáková A, Ju Sung Y, Tachmazidou I, Teumer A, Thorleifsson G, van der Harst P, Walker RW, Wang SR, Wild SH, Willems SM, Wong A, Zhang W, Albrecht E, Couto Alves A, Bakker SJL, Barlassina C, Bartz TM, Beilby J, Bellis C, Bergman RN, Bergmann S, Blangero J, Blüher M, Boerwinkle E, Bonnycastle LL, Bornstein SR, Bruinenberg M, Campbell H, Chen YDI, Chiang CWK, Chines PS, Collins FS, Cucca F, Cupples LA, D'Avila F, de Geus EJ.C, Dedoussis G, Dimitriou M, Döring A, Eriksson JG, Farmaki AE, Farrall M, Ferreira T, Fischer K, Forouhi NG, Friedrich N, Gjesing AP, Glorioso N, Graff M, Grallert H, Grarup N, Gräßler J, Grewal J, Hamsten A, Harder MN, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Havulinna AS, Heliövaara M, Hillege H, Hofman A, Holmen O, Homuth G, Hottenga JJ, Hui J, Husemoen LL, Hysi PG, Isaacs A, Ittermann T, Jalilzadeh S, James AL, Jørgensen T, Jousilahti P, Jula A, Marie Justesen J, Justice AE, Kähönen M, Karaleftheri M, Tee Khaw K, Keinanen-Kiukaanniemi SM, Kinnunen L, Knekt PB, Koistinen HA, Kolcic I, Kooner IK, Koskinen S, Kovacs P, Kyriakou T, Laitinen T, Langenberg C, Lewin AM, Lichtner P, Lindgren CM, Lindström J, Linneberg A, Lorbeer R, Lorentzon M, Luben R, Lyssenko V, Männistö S, Manunta P, Leach IM, McArdle WL, Mcknight B, Mohlke KL, Mihailov E, Milani L, Mills R, Montasser ME, Morris AP, Müller G, Musk AW, Narisu N, Ong KK, Oostra BA, Osmond C, Palotie A, Pankow JS, Paternoster L, Penninx BW, Pichler I, Pilia MG, Polašek O, Pramstaller PP, Raitakari OT, Rankinen T, Rao DC, Rayner NW, Ribel-Madsen R, Rice TK, Richards M, Ridker PM, Rivadeneira F, Ryan KA, Sanna S, Sarzynski MA, Scholtens S, Scott RA, Sebert S, Southam L, Sparsø TH, Steinthorsdottir V, Stirrups K, Stolk RP, Strauch K, Stringham HM, Swertz MA, Swift AJ, Tönjes A, Tsafantakis E, van der Most PJ, Van Vliet-Ostaptchouk JV, Vandenput L, Vartiainen E, Venturini C, Verweij N, Viikari JS, Vitart V, Vohl MC, Vonk JM, Waeber G, Widén E, Willemsen G, Wilsgaard T, Winkler TW, Wright AF, Yerges-Armstrong LM, Hua Zhao J, Carola Zillikens M, Boomsma DI, Bouchard C, Chambers JC, Chasman DI, Cusi D, Gansevoort RT, Gieger C, Hansen T, Hicks AA, Hu F, Hveem K, Jarvelin MR, Kajantie E, Kooner JS, Kuh D, Kuusisto J, Laakso M, Lakka TA, Lehtimäki T, Metspalu A, Njølstad I, Ohlsson C, Oldehinkel AJ, Palmer LJ, Pedersen O, Perola M, Peters A, Psaty BM, Puolijoki H, Rauramaa R, Rudan I, Salomaa V, Schwarz PEH, Shudiner AR, Smit JH, Sørensen TIA, Spector TD, Stefansson K, Stumvoll M, Tremblay A, Tuomilehto J, Uitterlinden AG, Uusitupa M, Völker U, Vollenweider P, Wareham NJ, Watkins H, Wilson JF, Zeggini E, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, van Duijn CM, Fox C, Groop LC, Heid IM, Hunter DJ, Kaplan RC, McCarthy MI, North KE, O'Connell JR, Schlessinger D, Thorsteinsdottir U, Strachan DP, Frayling T, Hirschhorn JN, Müller-Nurasyid M, Loos RJF. A principal component meta-analysis on multiple anthropometric traits identifies novel loci for body shape. Nat Commun 2016; 7:13357. [PMID: 27876822 PMCID: PMC5114527 DOI: 10.1038/ncomms13357] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/21/2016] [Indexed: 01/15/2023] Open
Abstract
Large consortia have revealed hundreds of genetic loci associated with anthropometric traits, one trait at a time. We examined whether genetic variants affect body shape as a composite phenotype that is represented by a combination of anthropometric traits. We developed an approach that calculates averaged PCs (AvPCs) representing body shape derived from six anthropometric traits (body mass index, height, weight, waist and hip circumference, waist-to-hip ratio). The first four AvPCs explain >99% of the variability, are heritable, and associate with cardiometabolic outcomes. We performed genome-wide association analyses for each body shape composite phenotype across 65 studies and meta-analysed summary statistics. We identify six novel loci: LEMD2 and CD47 for AvPC1, RPS6KA5/C14orf159 and GANAB for AvPC3, and ARL15 and ANP32 for AvPC4. Our findings highlight the value of using multiple traits to define complex phenotypes for discovery, which are not captured by single-trait analyses, and may shed light onto new pathways.
Collapse
Affiliation(s)
- Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Janina Jeff M.
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Jennifer L. Bragg-Gresham
- Kidney Epidemiology and Cost Center, Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jenny van Dongen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Tarunveer S. Ahluwalia
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Ledreborg Allé 34, DK-2820 Copenhagen, Denmark
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Niina Eklund
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Joel Eriksson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Tõnu Esko
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Nancy L. Heard-Costa
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Anne U. Jackson
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Eero Jokinen
- Hospital for Children and Adolescents, University of Helsinki, FI-00290 Helsinki, Finland
| | - Stavroula Kanoni
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kati Kristiansson
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), 1010 Lausanne, Switzerland
| | - Jari Lahti
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Institute of Behavioural Sciences, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Keri L. Monda
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- The Center for Observational Research, Amgen Inc., Thousand Oaks, California 91320-1799, USA
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Lu Qi
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Erika Salvi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Megan T. Smith
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ioanna Tachmazidou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | | | - Pim van der Harst
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute Netherlands-Netherlands Heart Institute, 3501 DG Utrecht, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sophie R. Wang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
| | - Sarah H. Wild
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Sara M. Willems
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Cristina Barlassina
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Claire Bellis
- Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4001, Australia
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research of Singapore, Singapore 138672, Singapore
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas 78520, USA
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Stefan R. Bornstein
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
| | - Marcel Bruinenberg
- University of Groningen, University Medical Center Groningen, The LifeLines Cohort Study, 9700 RB Groningen, The Netherlands
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Yii-Der Ida Chen
- Los Angeles BioMedical Resesarch Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | | | - L Adrienne Cupples
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
| | - Francesca D'Avila
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Eco J .C. de Geus
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, VU University Medical Center, 1081 BT Amsterdam, The Netherlands
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Maria Dimitriou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Johan G. Eriksson
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, FI-00014 Helsinki, Finland
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Anette Prior Gjesing
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari, 7100 Sassari, Italy
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Niels Grarup
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jürgen Gräßler
- Department of Medicine III, Pathobiochemistry, Technische Universitaet, 01307 Dresden, Germany
| | - Jagvir Grewal
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Anders Hamsten
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Atherosclerosis Research Unit, Karolinska Institutet, 17176 Stockholm 17176, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Marie Neergaard Harder
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Catharina A. Hartman
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Andrew Tym Hattersley
- Institue of Biomedical & Clinical Science, University of Exeter, Barrack Road, Exeter EX2 5DW, UK
| | - Aki S. Havulinna
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Hans Hillege
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Oddgeir Holmen
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennie Hui
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Population Health, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Lise Lotte Husemoen
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Aaron Isaacs
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Shapour Jalilzadeh
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Torben Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg, 9220 Aalborg, Denmark
- Research Centre for Prevention and Health, Capital Region of Denmark, DK2600 Glostrup, Denmark
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Johanne Marie Justesen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, FI-33521 Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, FI-33014 Tampere, Finland
| | | | - Kay Tee Khaw
- Clinical Gerontology Unit, Box 251, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Sirkka M. Keinanen-Kiukaanniemi
- Faculty of Medicine, Institute of Health Sciences, University of Oulu, Oulu F1-90014, Finland
- Unit of General Practice, Oulu University Hospital, Oulu FI-90029, Finland
| | - Leena Kinnunen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Paul B. Knekt
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Heikki A. Koistinen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital,, 00029 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | | | - Seppo Koskinen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
| | - Theodosios Kyriakou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Tomi Laitinen
- Kuopio University Hospital, 70029 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Alexandra M. Lewin
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Cecilia M. Lindgren
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- The Big Data Institute, University of Oxford, Oxford OX3 7LJ, UK
| | - Jaana Lindström
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, 2600 Glostrup, Denmark
| | - Roberto Lorbeer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mattias Lorentzon
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Robert Luben
- Strangeways Research Laboratory Wort's Causeway, Cambridge CB1 8RN, UK
| | - Valeriya Lyssenko
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Paolo Manunta
- Chair of Nephrology, Università Vita Salute San Raffaele and Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan 20139, Italy
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
| | - Barbara Mcknight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Divison of Public Health Sciences, Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | | | - May E. Montasser
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GA, UK
| | - Gabriele Müller
- Center for Evidence Based Healthcare, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, 01307, Germany
| | - Arthur W. Musk
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, West Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ben A. Oostra
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
- Massachusetts General Hospital, Center for Human Genetic Research, Psychiatric and Neurodevelopmental Genetics Unit, Boston, Massachusetts 02114, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota 55455-0381, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Brenda W. Penninx
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Irene Pichler
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Maria G. Pilia
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Ozren Polašek
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, General Central Hospital, 39100 Bolzano, Italy
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, FI-20520 Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Nigel W. Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
| | - Rasmus Ribel-Madsen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marcus Richards
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Kathy A. Ryan
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Sylvain Sebert
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Center For Life-Course Health Research, University of Oulu, FI-90014 Oulu, Finland
| | - Lorraine Southam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Thomas Hempel Sparsø
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | - Kathleen Stirrups
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ronald P. Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Heather M. Stringham
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | | | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Jana V. Van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Liesbeth Vandenput
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Erkki Vartiainen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Jorma S. Viikari
- Department of Medicine, University of Turku, FI-20521 Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
- School of Nutrition, Laval University, Québec, Québec, Canada G1V 0A6
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Gérard Waeber
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Tom Wilsgaard
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Laura M. Yerges-Armstrong
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Daniele Cusi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Ron T. Gansevoort
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Torben Hansen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Frank Hu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Kristian Hveem
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Marjo-Riitta Jarvelin
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Unit of Primary Care, Oulu University Hospital, 90029 OYS Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London W12 0NN, UK
- Faculty of Medicine, Center for Life Course Epidemiology, University of Oulu, P.O.Box 5000, FI-90014 Oulu, Finland
| | - Eero Kajantie
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
- Department of Obstetrics and Gynecology, MRC Oulu, Oulu University Hospital and University of Oulu, FI-90029 Oulu, Finland
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Physiology, Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, FI-33520 Tampere, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Inger Njølstad
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
- Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Albertine J. Oldehinkel
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Oluf Pedersen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Departments of Epidemiology and Health Services, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seatte, Washington 98101, USA
| | - Hannu Puolijoki
- South Ostrobothnia Central Hospital, Seinäjoki Fi-60220, Finland
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, 70211 Kuopio, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter E. H. Schwarz
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Dresden 01307, Germany
| | - Alan R. Shudiner
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Geriatric Research and Education Clinical Center, Vetrans Administration Medical Center, Baltimore, Maryland 21042, USA
| | - Jan H. Smit
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Thorkild I. A. Sørensen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, 2000 Frederiksberg, Denmark
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Angelo Tremblay
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
| | - Jaakko Tuomilehto
- Diabetes Prevention Unit, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Centre for Vascular Prevention, Danube-University Krems, 3500 Krems, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Matti Uusitupa
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
- Research Unit, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Goncalo R. Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Cornelia M. van Duijn
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
- Center for Medical Systems Biology, 2300 Leiden, The Netherlands
| | - Caroline Fox
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Leif C. Groop
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
- Finnish Institute for Molecular Medicine (FIMM), Helsinki University, 00014 Helsinki, Finland
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre, Oxford OX3 7LJ, UK
| | - Kari E. North
- Department of Epidemiology, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, USA
| | - Jeffrey R. O'Connell
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Timothy Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
- Metabolism Initiative, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
158
|
Chen Y, Fu F, Lin Y, Qiu L, Lu M, Zhang J, Qiu W, Yang P, Wu N, Huang M, Wang C. The precision relationships between eight GWAS-identified genetic variants and breast cancer in a Chinese population. Oncotarget 2016; 7:75457-75467. [PMID: 27705907 PMCID: PMC5342752 DOI: 10.18632/oncotarget.12255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/16/2016] [Indexed: 11/25/2022] Open
Abstract
Some of the new breast cancer susceptibility loci discovered in recent Genome-wide association studies (GWASs) have not been confirmed in Chinese populations. To determine whether eight novel Single-Nucleotide Polymorphisms (SNPs) have associations with breast cancer risk in women from southeast China, we conducted a case-control study of 1,156 breast cancer patients and 1,256 healthy controls. We first validated that the SNPs rs12922061, rs2290203, and rs2981578 were associated with overall breast cancer risk in southeast Chinese women, with the per-allele OR of 1.209 (95%CI: 1.064-1.372), 1.176 (95%CI: 1.048-1.320), and 0.852 (95%CI: 0.759-0.956), respectively. Rs12922061 and rs2290203 even passed the threshold for Bonferroni correction (P value: 0.00625). In stratified analysis, we found another three SNPs were significantly associated within different subgroups. However, after Bonferroni correction (P value: 0.000446), there were no statistically significant was observed. In gene-environment interaction analysis, we observed gene-environment interactions played a potential role of in the risk of breast cancer. These findings provide new insight into the associations between the genetic susceptibility and fine classifications of breast cancer. Based on these results, we encourage further large series studies and functional research to confirm these finding.
Collapse
Affiliation(s)
- Yazhen Chen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Fangmeng Fu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Yuxiang Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Lin Qiu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Minjun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Jiantang Zhang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Wei Qiu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Peidong Yang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Na Wu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Meng Huang
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian Province, 350001, China
| | - Chuan Wang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| |
Collapse
|
159
|
Matsson H, Söderhäll C, Einarsdottir E, Lamontagne M, Gudmundsson S, Backman H, Lindberg A, Rönmark E, Kere J, Sin D, Postma DS, Bossé Y, Lundbäck B, Klar J. Targeted high-throughput sequencing of candidate genes for chronic obstructive pulmonary disease. BMC Pulm Med 2016; 16:146. [PMID: 27835950 PMCID: PMC5106844 DOI: 10.1186/s12890-016-0309-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/06/2016] [Indexed: 12/13/2022] Open
Abstract
Background Reduced lung function in patients with chronic obstructive pulmonary disease (COPD) is likely due to both environmental and genetic factors. We report here a targeted high-throughput DNA sequencing approach to identify new and previously known genetic variants in a set of candidate genes for COPD. Methods Exons in 22 genes implicated in lung development as well as 61 genes and 10 genomic regions previously associated with COPD were sequenced using individual DNA samples from 68 cases with moderate or severe COPD and 66 controls matched for age, gender and smoking. Cases and controls were selected from the Obstructive Lung Disease in Northern Sweden (OLIN) studies. Results In total, 37 genetic variants showed association with COPD (p < 0.05, uncorrected). Several variants previously discovered to be associated with COPD from genetic genome-wide analysis studies were replicated using our sample. Two high-risk variants were followed-up for functional characterization in a large eQTL mapping study of 1,111 human lung specimens. The C allele of a synonymous variant, rs8040868, predicting a p.(S45=) in the gene for cholinergic receptor nicotinic alpha 3 (CHRNA3) was associated with COPD (p = 8.8 x 10−3). This association remained (p = 0.003 and OR = 1.4, 95 % CI 1.1-1.7) when analysing all available cases and controls in OLIN (n = 1,534). The rs8040868 variant is in linkage disequilibrium with rs16969968 previously associated with COPD and altered expression of the CHRNA5 gene. A follow-up analysis for detection of expression quantitative trait loci revealed that rs8040868-C was found to be significantly associated with a decreased expression of the nearby gene cholinergic receptor, nicotinic, alpha 5 (CHRNA5) in lung tissue. Conclusion Our data replicate previous result suggesting CHRNA5 as a candidate gene for COPD and rs8040868 as a risk variant for the development of COPD in the Swedish population. Electronic supplementary material The online version of this article (doi:10.1186/s12890-016-0309-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hans Matsson
- Department of Biosciences and Nutrition, Karolinska Institutet, 7-9, SE-141 83, Huddinge, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, 7-9, SE-141 83, Huddinge, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Elisabet Einarsdottir
- Department of Biosciences and Nutrition, Karolinska Institutet, 7-9, SE-141 83, Huddinge, Sweden.,Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Maxime Lamontagne
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada
| | - Sanna Gudmundsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Backman
- Department of Public Health and Clinical Medicine, Division of Occupational and Environmental Medicine, Umeå University, Umeå, Sweden
| | - Anne Lindberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Eva Rönmark
- Department of Public Health and Clinical Medicine, Division of Occupational and Environmental Medicine, Umeå University, Umeå, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, 7-9, SE-141 83, Huddinge, Sweden.,Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Don Sin
- The University of British Columbia Center for Heart Lung Innovation, St-Paul's Hospital, Vancouver, Canada
| | - Dirkje S Postma
- Center Groningen, GRIAC research institute, University of Groningen, Groningen, The Netherlands
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, Canada.,Department of Molecular Medicine, Laval University, Québec, Canada
| | - Bo Lundbäck
- Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Joakim Klar
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
160
|
Franconi F, Rosano G, Basili S, Montella A, Campesi I. Human cells involved in atherosclerosis have a sex. Int J Cardiol 2016; 228:983-1001. [PMID: 27915217 DOI: 10.1016/j.ijcard.2016.11.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/06/2016] [Indexed: 12/30/2022]
Abstract
The influence of sex has been largely described in cardiovascular diseases. Atherosclerosis is a complex process that involves many cell types such as vessel cells, immune cells and endothelial progenitor cells; however, many, if not all, studies do not report the sex of the cells. This review focuses on sex differences in human cells involved in the atherosclerotic process, emphasizing the role of sex hormones. Furthermore, we report sex differences and issues related to the processes that determine the fate of the cells such as apoptotic and autophagic mechanisms. The analysis of the data reveals that there are still many gaps in our knowledge regarding sex influences in atherosclerosis, largely for the cell types that have not been well studied, stressing the urgent need for a clear definition of experimental conditions and the inclusion of both sexes in preclinical studies.
Collapse
Affiliation(s)
- Flavia Franconi
- Assessorato alle Politiche per la Persona of Basilicata Region, Potenza, Italy; Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Rosano
- Cardiovascular and Cell Sciences Research Institute, St. George's University of London, United Kingdom
| | - Stefania Basili
- Department of Internal Medicine and Medical Specialties - Research Center on Gender and Evaluation and Promotion of Quality in Medicine (CEQUAM), Sapienza University of Rome, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Osilo, Italy.
| |
Collapse
|
161
|
Genome-Wide Interaction Analyses between Genetic Variants and Alcohol Consumption and Smoking for Risk of Colorectal Cancer. PLoS Genet 2016; 12:e1006296. [PMID: 27723779 PMCID: PMC5065124 DOI: 10.1371/journal.pgen.1006296] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 08/11/2016] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified many genetic susceptibility loci for colorectal cancer (CRC). However, variants in these loci explain only a small proportion of familial aggregation, and there are likely additional variants that are associated with CRC susceptibility. Genome-wide studies of gene-environment interactions may identify variants that are not detected in GWAS of marginal gene effects. To study this, we conducted a genome-wide analysis for interaction between genetic variants and alcohol consumption and cigarette smoking using data from the Colon Cancer Family Registry (CCFR) and the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO). Interactions were tested using logistic regression. We identified interaction between CRC risk and alcohol consumption and variants in the 9q22.32/HIATL1 (Pinteraction = 1.76×10−8; permuted p-value 3.51x10-8) region. Compared to non-/occasional drinking light to moderate alcohol consumption was associated with a lower risk of colorectal cancer among individuals with rs9409565 CT genotype (OR, 0.82 [95% CI, 0.74–0.91]; P = 2.1×10−4) and TT genotypes (OR,0.62 [95% CI, 0.51–0.75]; P = 1.3×10−6) but not associated among those with the CC genotype (p = 0.059). No genome-wide statistically significant interactions were observed for smoking. If replicated our suggestive finding of a genome-wide significant interaction between genetic variants and alcohol consumption might contribute to understanding colorectal cancer etiology and identifying subpopulations with differential susceptibility to the effect of alcohol on CRC risk. Alcohol consumption and smoking are associated with CRC risk. We performed a genome-wide analysis for interaction between genetic variants and alcohol consumption and cigarette smoking to identify potential new genetic regions associated with CRC. About 8,000 CRC cases and 8,800 controls were included in alcohol-related analysis and over 11,000 cases and 11,000 controls were involved in smoking-related analysis. We identified interaction between variants at 9q22.32/HIATL1 and alcohol consumption in relation to CRC risk (Pinteraction = 1.76×10−8). If replicated our suggested finding of the interaction between genetic variants and alcohol consumption might contribute to understanding colorectal cancer etiology and identifying subpopulations with differential susceptible to the effect of alcohol on CRC risk.
Collapse
|
162
|
Liu C, Kraja AT, Smith JA, Brody JA, Franceschini N, Bis JC, Rice K, Morrison AC, Lu Y, Weiss S, Guo X, Palmas W, Martin LW, Chen YDI, Surendran P, Drenos F, Cook JP, Auer PL, Chu AY, Giri A, Zhao W, Jakobsdottir J, Lin LA, Stafford JM, Amin N, Mei H, Yao J, Voorman A, Larson MG, Grove ML, Smith AV, Hwang SJ, Chen H, Huan T, Kosova G, Stitziel NO, Kathiresan S, Samani N, Schunkert H, Deloukas P, Li M, Fuchsberger C, Pattaro C, Gorski M, Kooperberg C, Papanicolaou GJ, Rossouw JE, Faul JD, Kardia SLR, Bouchard C, Raffel LJ, Uitterlinden AG, Franco OH, Vasan RS, O'Donnell CJ, Taylor KD, Liu K, Bottinger EP, Gottesman O, Daw EW, Giulianini F, Ganesh S, Salfati E, Harris TB, Launer LJ, Dörr M, Felix SB, Rettig R, Völzke H, Kim E, Lee WJ, Lee IT, Sheu WHH, Tsosie KS, Edwards DRV, Liu Y, Correa A, Weir DR, Völker U, Ridker PM, Boerwinkle E, Gudnason V, Reiner AP, van Duijn CM, Borecki IB, Edwards TL, Chakravarti A, Rotter JI, Psaty BM, Loos RJF, Fornage M, Ehret GB, Newton-Cheh C, Levy D, Chasman DI. Meta-analysis identifies common and rare variants influencing blood pressure and overlapping with metabolic trait loci. Nat Genet 2016; 48:1162-70. [PMID: 27618448 PMCID: PMC5320952 DOI: 10.1038/ng.3660] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 08/05/2016] [Indexed: 11/08/2022]
Abstract
Meta-analyses of association results for blood pressure using exome-centric single-variant and gene-based tests identified 31 new loci in a discovery stage among 146,562 individuals, with follow-up and meta-analysis in 180,726 additional individuals (total n = 327,288). These blood pressure-associated loci are enriched for known variants for cardiometabolic traits. Associations were also observed for the aggregation of rare and low-frequency missense variants in three genes, NPR1, DBH, and PTPMT1. In addition, blood pressure associations at 39 previously reported loci were confirmed. The identified variants implicate biological pathways related to cardiometabolic traits, vascular function, and development. Several new variants are inferred to have roles in transcription or as hubs in protein-protein interaction networks. Genetic risk scores constructed from the identified variants were strongly associated with coronary disease and myocardial infarction. This large collection of blood pressure-associated loci suggests new therapeutic strategies for hypertension, emphasizing a link with cardiometabolic risk.
Collapse
Affiliation(s)
- Chunyu Liu
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nora Franceschini
- Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Yingchang Lu
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stefan Weiss
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Walter Palmas
- Division of General Medicine, Columbia University Medical Center, New York, New York, USA
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Praveen Surendran
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Fotios Drenos
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Audrey Y Chu
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ayush Giri
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Li-An Lin
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jeanette M Stafford
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Hao Mei
- Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Arend Voorman
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Martin G Larson
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Shih-Jen Hwang
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Han Chen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tianxiao Huan
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Gulum Kosova
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Nathan O Stitziel
- Division of Cardiology, Department of Medicine and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sekar Kathiresan
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Nilesh Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Panos Deloukas
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Man Li
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christian Fuchsberger
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), Bolzano, Italy (affiliated with the University of Lübeck, Lübeck, Germany)
| | - Cristian Pattaro
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), Bolzano, Italy (affiliated with the University of Lübeck, Lübeck, Germany)
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - George J Papanicolaou
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jacques E Rossouw
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Claude Bouchard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Leslie J Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Ramachandran S Vasan
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Preventive Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Christopher J O'Donnell
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Cardiology Section, Department of Medicine, Boston Veterans Administration Healthcare, Boston, Massachusetts, USA
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Kiang Liu
- Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Erwin P Bottinger
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Omri Gottesman
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Santhi Ganesh
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elias Salfati
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, US National Institutes of Health, Bethesda, Maryland, USA
| | - Lenore J Launer
- Neuroepidemiology Section, National Institute on Aging, US National Institutes of Health, Bethesda, Maryland, USA
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Rainer Rettig
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Physiology, University of Greifswald, Greifswald, Germany
| | - Henry Völzke
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- DZD (German Center for Diabetes Research), site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Eric Kim
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Krystal S Tsosie
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Digna R Velez Edwards
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yongmei Liu
- Epidemiology and Prevention Center for Genomics and Personalized Medicine Research, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Todd L Edwards
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Health Services, University of Washington, Seattle, Washington, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Christopher Newton-Cheh
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
163
|
Abstract
Mammalian immune system is a complex amalgam of diverse cellular and noncellular components such as cytokines, receptors and co-receptors. FAM26F (family with sequence similarity 26, member F) is a recently identified tetraspanin-like membrane glycoprotein which is predicted to make homophilic interactions and potential synapses between several immune cells including CD4+, CD8+, NK, dendritic cells and macrophages. Various whole transcriptome analyses have demonstrated the differential expression of FAM26F in several bacterial, viral and parasitic infections, in certain pathophysiological conditions such as liver and heart transplantation, and in various cancers. The complete understanding of transcriptional regulation of FAM26F is in its infancy however it is up regulated by various stimulants such as polyI:C, LPS, INF gamma and TNF alpha, and via various proposed pathways including TLR3, TLR4 IFN-β and Dectin-1. These pathways can merge in STAT1 activation. The synergistic expression of FAM26F on both NK-cells and myeloid dendritic cells is required to activate NK-cells against tumors via its cytoplasmic tail, thus emphasizing therapeutic potential of FAM26F for NK sensitive tumors. Current review provides a comprehensive basis to propose that FAM26F expression level is at least a hallmark for IFN-γ-lead immune responses and thus can proficiently be regarded as an early diagnostic marker. Future investigation dissecting the role of FAM26F in activation of various immune cell populations in local amplification by cell-cell contact is crucial to provide the missing link imperative for elucidating the relevance of this protein in immune responses.
Collapse
Affiliation(s)
- Uzma Malik
- a Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB) , National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Aneela Javed
- a Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB) , National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| |
Collapse
|
164
|
Aminkeng F, Ross CJD, Rassekh SR, Hwang S, Rieder MJ, Bhavsar AP, Smith A, Sanatani S, Gelmon KA, Bernstein D, Hayden MR, Amstutz U, Carleton BC. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol 2016; 82:683-95. [PMID: 27197003 PMCID: PMC5338111 DOI: 10.1111/bcp.13008] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022] Open
Abstract
AIMS Anthracycline-induced cardiotoxicity (ACT) occurs in 57% of treated patients and remains an important limitation of anthracycline-based chemotherapy. In various genetic association studies, potential genetic risk markers for ACT have been identified. Therefore, we developed evidence-based clinical practice recommendations for pharmacogenomic testing to further individualize therapy based on ACT risk. METHODS We followed a standard guideline development process, including a systematic literature search, evidence synthesis and critical appraisal, and the development of clinical practice recommendations with an international expert group. RESULTS RARG rs2229774, SLC28A3 rs7853758 and UGT1A6 rs17863783 variants currently have the strongest and the most consistent evidence for association with ACT. Genetic variants in ABCC1, ABCC2, ABCC5, ABCB1, ABCB4, CBR3, RAC2, NCF4, CYBA, GSTP1, CAT, SULT2B1, POR, HAS3, SLC22A7, SCL22A17, HFE and NOS3 have also been associated with ACT, but require additional validation. We recommend pharmacogenomic testing for the RARG rs2229774 (S427L), SLC28A3 rs7853758 (L461L) and UGT1A6*4 rs17863783 (V209V) variants in childhood cancer patients with an indication for doxorubicin or daunorubicin therapy (Level B - moderate). Based on an overall risk stratification, taking into account genetic and clinical risk factors, we recommend a number of management options including increased frequency of echocardiogram monitoring, follow-up, as well as therapeutic options within the current standard of clinical practice. CONCLUSIONS Existing evidence demonstrates that genetic factors have the potential to improve the discrimination between individuals at higher and lower risk of ACT. Genetic testing may therefore support both patient care decisions and evidence development for an improved prevention of ACT.
Collapse
Affiliation(s)
- Folefac Aminkeng
- Centre for Molecular Medicine and Therapeutics, Department of Medical GeneticsUniversity of British ColumbiaVancouverBCCanada
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Colin J. D. Ross
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Shahrad R. Rassekh
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Pediatric Hematology/Oncology/BMT, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Soomi Hwang
- Faculty of Pharmaceutical SciencesUniversity of British ColumbiaVancouverBCCanada
| | | | - Amit P. Bhavsar
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
| | - Anne Smith
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Pharmaceutical Outcomes & Policy Innovations ProgrammeBC Children's HospitalVancouverBCCanada
| | - Shubhayan Sanatani
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
| | | | - Daniel Bernstein
- Department of Pediatrics, Division of CardiologyStanford UniversityStanfordCAUSA
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, Department of Medical GeneticsUniversity of British ColumbiaVancouverBCCanada
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Translational Laboratory in Genetic Medicine, National University of Singapore and Association for ScienceTechnology and Research (A*STAR)Singapore
| | - Ursula Amstutz
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Division of Translational Therapeutics, Department of PediatricsUniversity of British ColumbiaVancouverBCCanada
- University Institute of Clinical Chemistry, Inselspital Bern University Hospital and University of BernSwitzerland
| | - Bruce C. Carleton
- Child & Family Research InstituteUniversity of British ColumbiaVancouverBCCanada
- Pharmaceutical Outcomes & Policy Innovations ProgrammeBC Children's HospitalVancouverBCCanada
| | | |
Collapse
|
165
|
Zhu YP, Thomas GD, Hedrick CC. 2014 Jeffrey M. Hoeg Award Lecture: Transcriptional Control of Monocyte Development. Arterioscler Thromb Vasc Biol 2016; 36:1722-33. [PMID: 27386937 PMCID: PMC5828163 DOI: 10.1161/atvbaha.116.304054] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/24/2016] [Indexed: 01/01/2023]
Abstract
Monocytes and macrophages are key immune cells involved in the early progression of atherosclerosis. Transcription factors that control their development in the bone marrow are important therapeutic targets to control the numbers and functions of these cells in disease. This review highlights what is currently known about the transcription factors that are critical for monocyte development.
Collapse
Affiliation(s)
- Yanfang Peipei Zhu
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Graham D Thomas
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
166
|
Li S, Sun YN, Zhou YT, Zhang CL, Lu F, Liu J, Shang XM. Screening and identification of microRNA involved in unstable angina using gene-chip analysis. Exp Ther Med 2016; 12:2716-2722. [PMID: 27703515 DOI: 10.3892/etm.2016.3646] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/02/2016] [Indexed: 01/05/2023] Open
Abstract
Increasing evidence has suggested that microRNA (miRNA) may play a role in the pathogenesis of cardiovascular disease, which has led to a greater understanding of the complex pathophysiological processes underlying unstable angina (UA). The present study aimed to investigate changes in the miRNA expression profiles of patients with UA using gene-chip analysis, in order to further elucidate the pathogenesis of UA. Total RNA was extracted and purified from plasma samples collected from patients with UA and healthy controls. The samples underwent microarray analysis using an Exiqon miRCURY LNA™ microRNA Array. Differentially expressed miRNAs were identified by volcano plot filtering, and were validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). In addition, functional annotation of the differentially expressed miRNAs involved gene ontology analyses. Among the 212 miRNAs differentially expressed between the two groups, 82 were upregulated and 130 were downregulated. Notably, the results of the RT-qPCR were consistent with the gene-chip results. The miRNAs identified in the present study may be potential novel biomarkers for the prevention and early diagnosis of UA. Furthermore, the results of the present study suggested that UA occurs as a result of complex and dynamic processes regulated by numerous factors, including multiple miRNAs.
Collapse
Affiliation(s)
- Si Li
- Department of Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ya-Nan Sun
- Department of Cardiology, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yun-Tao Zhou
- Department of Cardiology, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Chun-Lai Zhang
- Department of Cardiology, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Feng Lu
- Department of Cardiology, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jia Liu
- Department of Cardiology, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xiao-Ming Shang
- Department of Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
167
|
Ferreira MAR, Jansen R, Willemsen G, Penninx B, Bain LM, Vicente CT, Revez JA, Matheson MC, Hui J, Tung JY, Baltic S, Le Souëf P, Montgomery GW, Martin NG, Robertson CF, James A, Thompson PJ, Boomsma DI, Hopper JL, Hinds DA, Werder RB, Phipps S. Gene-based analysis of regulatory variants identifies 4 putative novel asthma risk genes related to nucleotide synthesis and signaling. J Allergy Clin Immunol 2016; 139:1148-1157. [PMID: 27554816 DOI: 10.1016/j.jaci.2016.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Hundreds of genetic variants are thought to contribute to variation in asthma risk by modulating gene expression. Methods that increase the power of genome-wide association studies (GWASs) to identify risk-associated variants are needed. OBJECTIVE We sought to develop a method that aggregates the evidence for association with disease risk across expression quantitative trait loci (eQTLs) of a gene and use this approach to identify asthma risk genes. METHODS We developed a gene-based test and software package called EUGENE that (1) is applicable to GWAS summary statistics; (2) considers both cis- and trans-eQTLs; (3) incorporates eQTLs identified in different tissues; and (4) uses simulations to account for multiple testing. We applied this approach to 2 published asthma GWASs (combined n = 46,044) and used mouse studies to provide initial functional insights into 2 genes with novel genetic associations. RESULTS We tested the association between asthma and 17,190 genes that were found to have cis- and/or trans-eQTLs across 16 published eQTL studies. At an empirical FDR of 5%, 48 genes were associated with asthma risk. Of these, for 37, the association was driven by eQTLs located in established risk loci for allergic disease, including 6 genes not previously implicated in disease cause (eg, LIMS1, TINF2, and SAFB). The remaining 11 significant genes represent potential novel genetic associations with asthma. The association with 4 of these replicated in an independent GWAS: B4GALT3, USMG5, P2RY13, and P2RY14, which are genes involved in nucleotide synthesis or nucleotide-dependent cell activation. In mouse studies, P2ry13 and P2ry14-purinergic receptors activated by adenosine 5-diphosphate and UDP-sugars, respectively-were upregulated after allergen challenge, notably in airway epithelial cells, eosinophils, and neutrophils. Intranasal exposure with receptor agonists induced the release of IL-33 and subsequent eosinophil infiltration into the lungs. CONCLUSION We identified novel associations between asthma and eQTLs for 4 genes related to nucleotide synthesis/signaling and demonstrated the power of gene-based analyses of GWASs.
Collapse
Affiliation(s)
| | - Rick Jansen
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - Brenda Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Lisa M Bain
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Joana A Revez
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Melanie C Matheson
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, Australia; School of Population Health, University of Western Australia, Nedlands, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia; Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Nedlands, Australia
| | | | - Svetlana Baltic
- Institute for Respiratory Health, Harry Perkins Institute of Medical Research, Nedlands, Australia
| | - Peter Le Souëf
- School of Paediatrics and Child Health, Princess Margaret Hospital for Children, Subiaco, Australia
| | | | | | - Colin F Robertson
- Respiratory Medicine, Murdoch Children's Research Institute, Melbourne, Australia
| | - Alan James
- Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Nedlands, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia; Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Nedlands, Australia
| | - Philip J Thompson
- Institute for Respiratory Health, Harry Perkins Institute of Medical Research, Nedlands, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - John L Hopper
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | | | - Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Simon Phipps
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
168
|
Joshi AD, Andersson C, Buch S, Stender S, Noordam R, Weng LC, Weeke PE, Auer PL, Boehm B, Chen C, Choi H, Curhan G, Denny JC, De Vivo I, Eicher JD, Ellinghaus D, Folsom AR, Fuchs C, Gala M, Haessler J, Hofman A, Hu F, Hunter DJ, Janssen HL, Kang JH, Kooperberg C, Kraft P, Kratzer W, Lieb W, Lutsey PL, Murad SD, Nordestgaard BG, Pasquale LR, Reiner AP, Ridker PM, Rimm E, Rose LM, Shaffer CM, Schafmayer C, Tamimi RM, Uitterlinden AG, Völker U, Völzke H, Wakabayashi Y, Wiggs JL, Zhu J, Roden DM, Stricker BH, Tang W, Teumer A, Hampe J, Tybjærg-Hansen A, Chasman DI, Chan AT, Johnson AD. Four Susceptibility Loci for Gallstone Disease Identified in a Meta-analysis of Genome-Wide Association Studies. Gastroenterology 2016; 151:351-363.e28. [PMID: 27094239 PMCID: PMC4959966 DOI: 10.1053/j.gastro.2016.04.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS A genome-wide association study (GWAS) of 280 cases identified the hepatic cholesterol transporter ABCG8 as a locus associated with risk for gallstone disease, but findings have not been reported from any other GWAS of this phenotype. We performed a large-scale, meta-analysis of GWASs of individuals of European ancestry with available prior genotype data, to identify additional genetic risk factors for gallstone disease. METHODS We obtained per-allele odds ratio (OR) and standard error estimates using age- and sex-adjusted logistic regression models within each of the 10 discovery studies (8720 cases and 55,152 controls). We performed an inverse variance weighted, fixed-effects meta-analysis of study-specific estimates to identify single-nucleotide polymorphisms that were associated independently with gallstone disease. Associations were replicated in 6489 cases and 62,797 controls. RESULTS We observed independent associations for 2 single-nucleotide polymorphisms at the ABCG8 locus: rs11887534 (OR, 1.69; 95% confidence interval [CI], 1.54-1.86; P = 2.44 × 10(-60)) and rs4245791 (OR, 1.27; P = 1.90 × 10(-34)). We also identified and/or replicated associations for rs9843304 in TM4SF4 (OR, 1.12; 95% CI, 1.08-1.16; P = 6.09 × 10(-11)), rs2547231 in SULT2A1 (encodes a sulfoconjugation enzyme that acts on hydroxysteroids and cholesterol-derived sterol bile acids) (OR, 1.17; 95% CI, 1.12-1.21; P = 2.24 × 10(-10)), rs1260326 in glucokinase regulatory protein (OR, 1.12; 95% CI, 1.07-1.17; P = 2.55 × 10(-10)), and rs6471717 near CYP7A1 (encodes an enzyme that catalyzes conversion of cholesterol to primary bile acids) (OR, 1.11; 95% CI, 1.08-1.15; P = 8.84 × 10(-9)). Among individuals of African American and Hispanic American ancestry, rs11887534 and rs4245791 were associated positively with gallstone disease risk, whereas the association for the rs1260326 variant was inverse. CONCLUSIONS In this large-scale GWAS of gallstone disease, we identified 4 loci in genes that have putative functions in cholesterol metabolism and transport, and sulfonylation of bile acids or hydroxysteroids.
Collapse
Affiliation(s)
- Amit D. Joshi
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA,Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital Boston, MA,To whom correspondence should be addressed: Amit D. Joshi, MBBS, PhD, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA. Tel: +1 617 724 7558; Charlotte Andersson, MD, PhD, The Framingham Heart Study, 73 Mt Wayte Avenue, Framingham, Massachusetts 01702, USA. , Andrew T. Chan, MD, MPH, Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, GRJ-825C, Boston, Massachusetts 02114, USA. Tel:+1 617 724 0283; Fax: +1 617 726 3673; , Andrew D. Johnson, PhD, Division of Intramural Research, National Heart, Lung and Blood Institute, Cardiovascular Epidemiology and Human Genomics Branch, The Framingham Heart Study, 73 Mt. Wayte Ave., Suite #2, Framingham, MA, 01702, USA. Tel: +1 508 663 4082; Fax: +1 508 626 1262;
| | - Charlotte Andersson
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts.
| | - Stephan Buch
- Medical Department 1, University Hospital Dresden, TU Dresden, Dresden Germany
| | - Stefan Stender
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Raymond Noordam
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Lu-Chen Weng
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN
| | - Peter E. Weeke
- Department of Medicine, Vanderbilt University, Nashville, TN,Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Paul L. Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin, Milwaukee,Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Bernhard Boehm
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Constance Chen
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA
| | - Hyon Choi
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Gary Curhan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Joshua C. Denny
- Department of Medicine, Vanderbilt University, Nashville, TN,Department of Biomedical Informatics, Vanderbilt University, Nashville, TN
| | - Immaculata De Vivo
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - John D. Eicher
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA,Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN
| | - Charles Fuchs
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Manish Gala
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Frank Hu
- Department of Epidemiology, Harvard School of Public Health, Boston, MA,Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - David J. Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Harry L.A. Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, the Netherlands,Toronto Centre for Liver Disease, Toronto Western and General Hospital, University Health Network, Toronto, Canada
| | - Jae H. Kang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Wolfgang Kratzer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, Christian Albrechts Universität Kiel, Niemannsweg 11, Kiel, Germany
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN
| | - Sarwa Darwish Murad
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, the Netherlands
| | - Børge G. Nordestgaard
- The Copenhagen General Population Study and,Department of Clinical Biochemistry, Herlev Hospital, Herlev Denmark,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louis R. Pasquale
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Paul M Ridker
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Eric Rimm
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA,Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Lynda M. Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | | | - Clemens Schafmayer
- Department of General, Abdominal, Thoracic and Transplantation Surgery, University of Kiel, Kiel, Germany
| | - Rulla M. Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany,German Center for Cardiovascular Research, Partner Site Greifswald,German Center for Diabetes Research, Site Greifswald
| | - Yoshiyuki Wakabayashi
- The National Heart, Lung, and Blood Institute, DNA Sequencing Core Laboratory, Bethesda, MD
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA
| | - Jun Zhu
- The National Heart, Lung, and Blood Institute, DNA Sequencing Core Laboratory, Bethesda, MD
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Bruno H. Stricker
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, TU Dresden, Dresden Germany
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark,Department of Clinical Biochemistry, Herlev Hospital, Herlev Denmark
| | - Daniel I. Chasman
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Andrew T. Chan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,To whom correspondence should be addressed: Amit D. Joshi, MBBS, PhD, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA. Tel: +1 617 724 7558; Charlotte Andersson, MD, PhD, The Framingham Heart Study, 73 Mt Wayte Avenue, Framingham, Massachusetts 01702, USA. , Andrew T. Chan, MD, MPH, Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, GRJ-825C, Boston, Massachusetts 02114, USA. Tel:+1 617 724 0283; Fax: +1 617 726 3673; , Andrew D. Johnson, PhD, Division of Intramural Research, National Heart, Lung and Blood Institute, Cardiovascular Epidemiology and Human Genomics Branch, The Framingham Heart Study, 73 Mt. Wayte Ave., Suite #2, Framingham, MA, 01702, USA. Tel: +1 508 663 4082; Fax: +1 508 626 1262;
| | - Andrew D. Johnson
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA,Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA,To whom correspondence should be addressed: Amit D. Joshi, MBBS, PhD, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA. Tel: +1 617 724 7558; Charlotte Andersson, MD, PhD, The Framingham Heart Study, 73 Mt Wayte Avenue, Framingham, Massachusetts 01702, USA. , Andrew T. Chan, MD, MPH, Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, GRJ-825C, Boston, Massachusetts 02114, USA. Tel:+1 617 724 0283; Fax: +1 617 726 3673; , Andrew D. Johnson, PhD, Division of Intramural Research, National Heart, Lung and Blood Institute, Cardiovascular Epidemiology and Human Genomics Branch, The Framingham Heart Study, 73 Mt. Wayte Ave., Suite #2, Framingham, MA, 01702, USA. Tel: +1 508 663 4082; Fax: +1 508 626 1262;
| |
Collapse
|
169
|
Richard AC, Peters JE, Lee JC, Vahedi G, Schäffer AA, Siegel RM, Lyons PA, Smith KGC. Targeted genomic analysis reveals widespread autoimmune disease association with regulatory variants in the TNF superfamily cytokine signalling network. Genome Med 2016; 8:76. [PMID: 27435189 PMCID: PMC4952362 DOI: 10.1186/s13073-016-0329-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/21/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Tumour necrosis factor (TNF) superfamily cytokines and their receptors regulate diverse immune system functions through a common set of signalling pathways. Genetic variants in and expression of individual TNF superfamily cytokines, receptors and signalling proteins have been associated with autoimmune and inflammatory diseases, but their interconnected biology has been largely unexplored. METHODS We took a hypothesis-driven approach using available genome-wide datasets to identify genetic variants regulating gene expression in the TNF superfamily cytokine signalling network and the association of these variants with autoimmune and autoinflammatory disease. Using paired gene expression and genetic data, we identified genetic variants associated with gene expression, expression quantitative trait loci (eQTLs), in four peripheral blood cell subsets. We then examined whether eQTLs were dependent on gene expression level or the presence of active enhancer chromatin marks. Using these eQTLs as genetic markers of the TNF superfamily signalling network, we performed targeted gene set association analysis in eight autoimmune and autoinflammatory disease genome-wide association studies. RESULTS Comparison of TNF superfamily network gene expression and regulatory variants across four leucocyte subsets revealed patterns that differed between cell types. eQTLs for genes in this network were not dependent on absolute gene expression levels and were not enriched for chromatin marks of active enhancers. By examining autoimmune disease risk variants among our eQTLs, we found that risk alleles can be associated with either increased or decreased expression of co-stimulatory TNF superfamily cytokines, receptors or downstream signalling molecules. Gene set disease association analysis revealed that eQTLs for genes in the TNF superfamily pathway were associated with six of the eight autoimmune and autoinflammatory diseases examined, demonstrating associations beyond single genome-wide significant hits. CONCLUSIONS This systematic analysis of the influence of regulatory genetic variants in the TNF superfamily network reveals widespread and diverse roles for these cytokines in susceptibility to a number of immune-mediated diseases.
Collapse
Affiliation(s)
- Arianne C. Richard
- />Department of Medicine and Cambridge Institute for Medical Research, The University of Cambridge, Box 139, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
- />Autoimmunity Branch, National Institute for Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - James E. Peters
- />Department of Medicine and Cambridge Institute for Medical Research, The University of Cambridge, Box 139, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - James C. Lee
- />Department of Medicine and Cambridge Institute for Medical Research, The University of Cambridge, Box 139, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Golnaz Vahedi
- />Department of Genetics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Alejandro A. Schäffer
- />Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD 20894 USA
| | - Richard M. Siegel
- />Autoimmunity Branch, National Institute for Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Paul A. Lyons
- />Department of Medicine and Cambridge Institute for Medical Research, The University of Cambridge, Box 139, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Kenneth G. C. Smith
- />Department of Medicine and Cambridge Institute for Medical Research, The University of Cambridge, Box 139, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| |
Collapse
|
170
|
Miller JRC, Lo KK, Andre R, Hensman Moss DJ, Träger U, Stone TC, Jones L, Holmans P, Plagnol V, Tabrizi SJ. RNA-Seq of Huntington's disease patient myeloid cells reveals innate transcriptional dysregulation associated with proinflammatory pathway activation. Hum Mol Genet 2016; 25:2893-2904. [PMID: 27170315 PMCID: PMC5181590 DOI: 10.1093/hmg/ddw142] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022] Open
Abstract
Innate immune activation beyond the central nervous system is emerging as a vital component of the pathogenesis of neurodegeneration. Huntington's disease (HD) is a fatal neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The systemic innate immune system is thought to act as a modifier of disease progression; however, the molecular mechanisms remain only partially understood. Here we use RNA-sequencing to perform whole transcriptome analysis of primary monocytes from thirty manifest HD patients and thirty-three control subjects, cultured with and without a proinflammatory stimulus. In contrast with previous studies that have required stimulation to elicit phenotypic abnormalities, we demonstrate significant transcriptional differences in HD monocytes in their basal, unstimulated state. This includes previously undetected increased resting expression of genes encoding numerous proinflammatory cytokines, such as IL6 Further pathway analysis revealed widespread resting enrichment of proinflammatory functional gene sets, while upstream regulator analysis coupled with Western blotting suggests that abnormal basal activation of the NFĸB pathway plays a key role in mediating these transcriptional changes. That HD myeloid cells have a proinflammatory phenotype in the absence of stimulation is consistent with a priming effect of mutant huntingtin, whereby basal dysfunction leads to an exaggerated inflammatory response once a stimulus is encountered. These data advance our understanding of mutant huntingtin pathogenesis, establish resting myeloid cells as a key source of HD immune dysfunction, and further demonstrate the importance of systemic immunity in the potential treatment of HD and the wider study of neurodegeneration.
Collapse
Affiliation(s)
- James R C Miller
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kitty K Lo
- UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Ralph Andre
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Davina J Hensman Moss
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ulrike Träger
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Timothy C Stone
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Vincent Plagnol
- UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| |
Collapse
|
171
|
Polley S, Cipriani V, Khan JC, Shahid H, Moore AT, Yates JRW, Hollox EJ. Analysis of copy number variation at DMBT1 and age-related macular degeneration. BMC MEDICAL GENETICS 2016; 17:44. [PMID: 27416785 PMCID: PMC4946147 DOI: 10.1186/s12881-016-0311-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 07/07/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND DMBT1 is a gene that shows extensive copy number variation (CNV) that alters the number of bacteria-binding domains in the protein and has been shown to activate the complement pathway. It lies next to the ARMS2/HTRA1 genes in a region of chromosome 10q26, where single nucleotide variants have been strongly associated with age-related macular degeneration (AMD), the commonest cause of blindness in Western populations. Complement activation is thought to be a key factor in the pathogenesis of this condition. We sought to investigate whether DMBT1 CNV plays any role in the susceptibility to AMD. METHODS We analysed long-range linkage disequilibrium of DMBT1 CNV1 and CNV2 with flanking single nucleotide polymorphisms (SNPs) using our previously published CNV and HapMap Phase 3 SNP data in the CEPH Europeans from Utah (CEU). We then typed a large cohort of 860 AMD patients and 419 examined age-matched controls for copy number at DMBT1 CNV1 and CNV2 and combined these data with copy numbers from a further 480 unexamined controls. RESULTS We found weak linkage disequilibrium between DMBT1 CNV1 and CNV2 with the SNPs rs1474526 and rs714816 in the HTRA1/ARMS2 region. By directly analysing copy number variation, we found no evidence of association of CNV1 or CNV2 with AMD. CONCLUSIONS We have shown that copy number variation at DMBT1 does not affect risk of developing age-related macular degeneration and can therefore be ruled out from future studies investigating the association of structural variation at 10q26 with AMD.
Collapse
Affiliation(s)
- Shamik Polley
- Department of Genetics, University of Leicester, Leicester, UK
| | - Valentina Cipriani
- UCL Institute of Ophthalmology, University College London, London, UK
- UCL Genetics Institute, University College London, London, UK
- Moorfields Eye Hospital, London, UK
| | - Jane C Khan
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Australia
- Department of Ophthalmology, Royal Perth Hospital, Perth, Australia
| | - Humma Shahid
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
- Department of Ophthamology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anthony T Moore
- UCL Institute of Ophthalmology, University College London, London, UK
- Moorfields Eye Hospital, London, UK
- Department of Ophthalmology UCSF Medical School, San Francisco, USA
| | - John R W Yates
- UCL Institute of Ophthalmology, University College London, London, UK
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Edward J Hollox
- Department of Genetics, University of Leicester, Leicester, UK.
| |
Collapse
|
172
|
Revez JA, Matheson MC, Hui J, Baltic S, James A, Upham JW, Dharmage S, Thompson PJ, Martin NG, Hopper JL, Ferreira MAR. Identification of STOML2 as a putative novel asthma risk gene associated with IL6R. Allergy 2016; 71:1020-30. [PMID: 26932604 DOI: 10.1111/all.12869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Functional variants in the interleukin-6 receptor gene (IL6R) are associated with asthma risk. We hypothesized that genes co-expressed with IL6R might also be regulated by genetic polymorphisms that are associated with asthma risk. The aim of this study was to identify such genes. METHODS To identify genes whose expression was correlated with that of IL6R, we analyzed gene expression levels generated for 373 human lymphoblastoid cell lines by the Geuvadis consortium and for 38 hematopoietic cell types by the Differentiation Map Portal (DMAP) project. Genes correlated with IL6R were then screened for nearby single nucleotide polymorphisms (SNPs) that were significantly associated with both variation in gene expression levels (eSNPs) and asthma risk. RESULTS We identified 90 genes with expression levels correlated with those of IL6R and that also had a nearby eSNP associated with disease risk in a published asthma GWAS (N = 20 776). For 16 (18%) genes, the association between the eSNP and asthma risk replicated with the same direction of effect in a further independent published asthma GWAS (N = 27 378). Among the top replicated associations (FDR < 0.05) were eSNPs for four known (IL18R1, IL18RAP, BCL6, and STAT6) and one putative novel asthma risk gene, stomatin-like protein 2 (STOML2). The expression of STOML2 was negatively correlated with IL6R, while eSNPs that increased the expression of STOML2 were associated with an increased asthma risk. CONCLUSION The expression of STOML2, a gene that plays a key role in mitochondrial function and T-cell activation, is associated with both IL-6 signaling and asthma risk.
Collapse
Affiliation(s)
- J. A. Revez
- QIMR Berghofer Medical Research Institute; Brisbane Qld Australia
| | - M. C. Matheson
- Melbourne School of Population and Global Health; The University of Melbourne; Melbourne Vic. Australia
| | - J. Hui
- PathWest Laboratory Medicine of Western Australia (WA); Nedlands WA Australia
- School of Population Health; The University of WA; Nedlands WA Australia
- School of Pathology and Laboratory Medicine; The University of WA; Nedlands WA Australia
- Busselton Population Medical Research Institute; Sir Charles Gairdner Hospital; Perth WA Australia
| | - S. Baltic
- Institute for Respiratory Health; University of WA; Perth WA Australia
| | - A. James
- Busselton Population Medical Research Institute; Sir Charles Gairdner Hospital; Perth WA Australia
- School of Medicine and Pharmacology; University of Western Australia; Nedlands WA Australia
- Department of Pulmonary Physiology; West Australian Sleep Disorders Research Institute; Nedlands WA Australia
| | - J. W. Upham
- School of Medicine; Translational Research Institute; The University of Queensland; Brisbane Qld Australia
| | - S. Dharmage
- Melbourne School of Population and Global Health; The University of Melbourne; Melbourne Vic. Australia
| | - P. J. Thompson
- Institute for Respiratory Health; University of WA; Perth WA Australia
- School of Medicine and Pharmacology; University of Western Australia; Nedlands WA Australia
| | - N. G. Martin
- QIMR Berghofer Medical Research Institute; Brisbane Qld Australia
| | - J. L. Hopper
- Melbourne School of Population and Global Health; The University of Melbourne; Melbourne Vic. Australia
| | | | | |
Collapse
|
173
|
Nurnberg ST, Zhang H, Hand NJ, Bauer RC, Saleheen D, Reilly MP, Rader DJ. From Loci to Biology: Functional Genomics of Genome-Wide Association for Coronary Disease. Circ Res 2016; 118:586-606. [PMID: 26892960 DOI: 10.1161/circresaha.115.306464] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies have provided a rich collection of ≈ 58 coronary artery disease (CAD) loci that suggest the existence of previously unsuspected new biology relevant to atherosclerosis. However, these studies only identify genomic loci associated with CAD, and many questions remain even after a genomic locus is definitively implicated, including the nature of the causal variant(s) and the causal gene(s), as well as the directionality of effect. There are several tools that can be used for investigation of the functional genomics of these loci, and progress has been made on a limited number of novel CAD loci. New biology regarding atherosclerosis and CAD will be learned through the functional genomics of these loci, and the hope is that at least some of these new pathways relevant to CAD pathogenesis will yield new therapeutic targets for the prevention and treatment of CAD.
Collapse
Affiliation(s)
- Sylvia T Nurnberg
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Hanrui Zhang
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nicholas J Hand
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Robert C Bauer
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Danish Saleheen
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Muredach P Reilly
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| | - Daniel J Rader
- From the Division of Translational Medicine and Human Genetics, Department of Medicine (S.T.N., R.C.B., D.J.R.), Penn Cardiovascular Institute, Department of Medicine (H.Z., M.P.R., D.J.R.), Department of Genetics (N.J.H., D.J.R.), and Department of Biostatistics and Epidemiology (D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
174
|
Han MR, Long J, Choi JY, Low SK, Kweon SS, Zheng Y, Cai Q, Shi J, Guo X, Matsuo K, Iwasaki M, Shen CY, Kim MK, Wen W, Li B, Takahashi A, Shin MH, Xiang YB, Ito H, Kasuga Y, Noh DY, Matsuda K, Park MH, Gao YT, Iwata H, Tsugane S, Park SK, Kubo M, Shu XO, Kang D, Zheng W. Genome-wide association study in East Asians identifies two novel breast cancer susceptibility loci. Hum Mol Genet 2016; 25:3361-3371. [PMID: 27354352 DOI: 10.1093/hmg/ddw164] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/04/2016] [Accepted: 05/20/2016] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is one of the most common malignancies among women worldwide. Genetic factors have been shown to play an important role in breast cancer aetiology. We conducted a two-stage genome-wide association study (GWAS) including 14 224 cases and 14 829 controls of East Asian women to search for novel genetic susceptibility loci for breast cancer. Single nucleotide polymorphisms (SNPs) in two loci were found to be associated with breast cancer risk at the genome-wide significance level. The first locus, represented by rs12118297 at 1p22.3 (near the LMO4 gene), was associated with breast cancer risk with odds ratio (OR) and (95% confidence interval (CI)) of 0.91 (0.88-0.94) and a P-value of 4.48 × 10- 8 This association was replicated in another study, DRIVE GAME-ON Consortium, including 16 003 cases and 41 335 controls of European ancestry (OR = 0.95, 95% CI = 0.91-0.99, P-value = 0.019). The second locus, rs16992204 at 21q22.12 (near the LINC00160 gene), was associated with breast cancer risk with OR (95% CI) of 1.13 (1.07-1.18) and a P-value of 4.63 × 10 - 8 The risk allele frequency for this SNP is zero in European-ancestry populations in 1000 Genomes Project and thus its association with breast cancer risk cannot be assessed in DRIVE GAME-ON Consortium. Functional annotation using the ENCODE data indicates that rs12118297 might be located in a repressed element and locus 21q22.12 may affect breast cancer risk through regulating LINC00160 expressions and interaction with oestrogen receptor signalling. Our findings provide additional insights into the genetics of breast cancer.
Collapse
Affiliation(s)
- Mi-Ryung Han
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jirong Long
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Ji-Yeob Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Siew-Kee Low
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama 351-0198, Japan
| | - Sun-Seog Kweon
- Department of Preventive Medicine, Chonnam National University Medical School, Gwangju 61469, South Korea.,Jeonnam Regional Cancer Center, Chonnam National University Hwasun Hospital, Hwasun 58128, South Korea
| | - Ying Zheng
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Qiuyin Cai
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jiajun Shi
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Xingyi Guo
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Keitaro Matsuo
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan.,Department of Epidemiology, Nagoya University Graduates School of Medicine, Nagoya 464-8681, Japan
| | - Motoki Iwasaki
- Epidemiology Division, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo 104-0045, Japan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Taiwan Biobank, Academia Sinica, Taipei 115, Taiwan.,College of Public Health, China Medical University, Taichung 404, Taiwan
| | - Mi Kyung Kim
- Division of Cancer Epidemiology and Management, National Cancer Center, Gyeonggi-do 10408, South Korea
| | - Wanqing Wen
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama 351-0198, Japan
| | - Min-Ho Shin
- Department of Preventive Medicine, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai 200032, China
| | - Hidemi Ito
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Yoshio Kasuga
- Department of Surgery, Nagano Matsushiro General Hospital, Nagano 381-1231, Japan
| | - Dong-Young Noh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, South Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Koichi Matsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, the University of Tokyo, Tokyo 108-8639, Japan
| | - Min Ho Park
- Department of Surgery, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai 200032, China
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Central Hospital, Nagoya 464-8681, Japan
| | - Shoichiro Tsugane
- Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo 104-0045, Japan
| | - Sue K Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, South Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama 351-0198, Japan
| | - Xiao-Ou Shu
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Daehee Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, South Korea.,Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Wei Zheng
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| |
Collapse
|
175
|
Blauwendraat C, Francescatto M, Gibbs JR, Jansen IE, Simón-Sánchez J, Hernandez DG, Dillman AA, Singleton AB, Cookson MR, Rizzu P, Heutink P. Comprehensive promoter level expression quantitative trait loci analysis of the human frontal lobe. Genome Med 2016; 8:65. [PMID: 27287230 PMCID: PMC4903003 DOI: 10.1186/s13073-016-0320-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/19/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Expression quantitative trait loci (eQTL) analysis is a powerful method to detect correlations between gene expression and genomic variants and is widely used to interpret the biological mechanism underlying identified genome wide association studies (GWAS) risk loci. Numerous eQTL studies have been performed on different cell types and tissues of which the majority has been based on microarray technology. METHODS We present here an eQTL analysis based on cap analysis gene expression sequencing (CAGEseq) data created from human postmortem frontal lobe tissue combined with genotypes obtained through genotyping arrays, exome sequencing, and CAGEseq. Using CAGEseq as an expression profiling technique combined with these different genotyping techniques allows measurement of the molecular effect of variants on individual transcription start sites and increases the resolution of eQTL analysis by also including the non-annotated parts of the genome. RESULTS We identified 2410 eQTLs and show that non-coding transcripts are more likely to contain an eQTL than coding transcripts, in particular antisense transcripts. We provide evidence for how previously identified GWAS loci for schizophrenia (NRGN), Parkinson's disease, and Alzheimer's disease (PARK16 and MAPT loci) could increase the risk for disease at a molecular level. Furthermore, we demonstrate that CAGEseq improves eQTL analysis because variants obtained from CAGEseq are highly enriched for having a functional effect and thus are an efficient method towards the identification of causal variants. CONCLUSION Our data contain both coding and non-coding transcripts and has the added value that we have identified eQTLs for variants directly adjacent to TSS. Future eQTL studies would benefit from combining CAGEseq with RNA sequencing for a more complete interpretation of the transcriptome and increased understanding of eQTL signals.
Collapse
Affiliation(s)
- Cornelis Blauwendraat
- Applied Genomics for Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Margherita Francescatto
- Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - J Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging (NIA), Bethesda, Maryland, USA.,Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Iris E Jansen
- Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Clinical Genetics, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Javier Simón-Sánchez
- Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging (NIA), Bethesda, Maryland, USA
| | - Allissa A Dillman
- Laboratory of Neurogenetics, National Institute on Aging (NIA), Bethesda, Maryland, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging (NIA), Bethesda, Maryland, USA
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging (NIA), Bethesda, Maryland, USA
| | - Patrizia Rizzu
- Applied Genomics for Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Peter Heutink
- Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Clinical Genetics, VU University Medical Center (VUmc), Amsterdam, The Netherlands.,Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
176
|
Ghiassian SD, Menche J, Chasman DI, Giulianini F, Wang R, Ricchiuto P, Aikawa M, Iwata H, Müller C, Zeller T, Sharma A, Wild P, Lackner K, Singh S, Ridker PM, Blankenberg S, Barabási AL, Loscalzo J. Endophenotype Network Models: Common Core of Complex Diseases. Sci Rep 2016; 6:27414. [PMID: 27278246 PMCID: PMC4899691 DOI: 10.1038/srep27414] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/19/2016] [Indexed: 02/03/2023] Open
Abstract
Historically, human diseases have been differentiated and categorized based on the organ system in which they primarily manifest. Recently, an alternative view is emerging that emphasizes that different diseases often have common underlying mechanisms and shared intermediate pathophenotypes, or endo(pheno)types. Within this framework, a specific disease’s expression is a consequence of the interplay between the relevant endophenotypes and their local, organ-based environment. Important examples of such endophenotypes are inflammation, fibrosis, and thrombosis and their essential roles in many developing diseases. In this study, we construct endophenotype network models and explore their relation to different diseases in general and to cardiovascular diseases in particular. We identify the local neighborhoods (module) within the interconnected map of molecular components, i.e., the subnetworks of the human interactome that represent the inflammasome, thrombosome, and fibrosome. We find that these neighborhoods are highly overlapping and significantly enriched with disease-associated genes. In particular they are also enriched with differentially expressed genes linked to cardiovascular disease (risk). Finally, using proteomic data, we explore how macrophage activation contributes to our understanding of inflammatory processes and responses. The results of our analysis show that inflammatory responses initiate from within the cross-talk of the three identified endophenotypic modules.
Collapse
Affiliation(s)
- Susan Dina Ghiassian
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jörg Menche
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Theoretical Physics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ruisheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Piero Ricchiuto
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Müller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Tania Zeller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Amitabh Sharma
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philipp Wild
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Lübeck/Kiel, Hamburg, Germany.,Preventive Cardiology and Preventive Medicine, Dept. of Medicine 2, University Medical Center Mainz, Mainz, Germany.,Clinical Epidemiology, Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Karl Lackner
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Lübeck/Kiel, Hamburg, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Sasha Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stefan Blankenberg
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Albert-László Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Theoretical Physics, Budapest University of Technology and Economics, Budapest, Hungary.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Center for Network Science, Central European University, Budapest, Hungary
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
177
|
Müller C, Schillert A, Röthemeier C, Trégouët DA, Proust C, Binder H, Pfeiffer N, Beutel M, Lackner KJ, Schnabel RB, Tiret L, Wild PS, Blankenberg S, Zeller T, Ziegler A. Removing Batch Effects from Longitudinal Gene Expression - Quantile Normalization Plus ComBat as Best Approach for Microarray Transcriptome Data. PLoS One 2016; 11:e0156594. [PMID: 27272489 PMCID: PMC4896498 DOI: 10.1371/journal.pone.0156594] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 05/17/2016] [Indexed: 12/13/2022] Open
Abstract
Technical variation plays an important role in microarray-based gene expression studies, and batch effects explain a large proportion of this noise. It is therefore mandatory to eliminate technical variation while maintaining biological variability. Several strategies have been proposed for the removal of batch effects, although they have not been evaluated in large-scale longitudinal gene expression data. In this study, we aimed at identifying a suitable method for batch effect removal in a large study of microarray-based longitudinal gene expression. Monocytic gene expression was measured in 1092 participants of the Gutenberg Health Study at baseline and 5-year follow up. Replicates of selected samples were measured at both time points to identify technical variability. Deming regression, Passing-Bablok regression, linear mixed models, non-linear models as well as ReplicateRUV and ComBat were applied to eliminate batch effects between replicates. In a second step, quantile normalization prior to batch effect correction was performed for each method. Technical variation between batches was evaluated by principal component analysis. Associations between body mass index and transcriptomes were calculated before and after batch removal. Results from association analyses were compared to evaluate maintenance of biological variability. Quantile normalization, separately performed in each batch, combined with ComBat successfully reduced batch effects and maintained biological variability. ReplicateRUV performed perfectly in the replicate data subset of the study, but failed when applied to all samples. All other methods did not substantially reduce batch effects in the replicate data subset. Quantile normalization plus ComBat appears to be a valuable approach for batch correction in longitudinal gene expression data.
Collapse
Affiliation(s)
- Christian Müller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, 20246, Germany
- Institute of Medical Biometry and Statistics, University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, 23562, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
| | - Arne Schillert
- Institute of Medical Biometry and Statistics, University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, 23562, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
| | - Caroline Röthemeier
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, 20246, Germany
| | - David-Alexandre Trégouët
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche(UMR) enSanté 1166, F-75013, Paris, France
- Institute for Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, F-75013, Paris, France
| | - Carole Proust
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche(UMR) enSanté 1166, F-75013, Paris, France
- Institute for Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, F-75013, Paris, France
| | - Harald Binder
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, 55131, Germany
| | - Manfred Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Karl J. Lackner
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Renate B. Schnabel
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, 20246, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
| | - Laurence Tiret
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche(UMR) enSanté 1166, F-75013, Paris, France
- Institute for Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, F-75013, Paris, France
| | - Philipp S. Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center Mainz, Mainz, 55131, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, 55131, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Rhine Main, Mainz, 55131, Germany
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, 20246, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, 20246, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
| | - Andreas Ziegler
- Institute of Medical Biometry and Statistics, University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, 23562, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Hamburg, Lübeck, Kiel, 20246, Germany
- Center for Clinical Trials, University of Lübeck, Lübeck, 23562, Germany
- * E-mail:
| |
Collapse
|
178
|
Yucesoy B, Talzhanov Y, Michael Barmada M, Johnson VJ, Kashon ML, Baron E, Wilson NW, Frye B, Wang W, Fluharty K, Gharib R, Meade J, Germolec D, Luster MI, Nedorost S. Association of MHC region SNPs with irritant susceptibility in healthcare workers. J Immunotoxicol 2016; 13:738-44. [PMID: 27258892 DOI: 10.3109/1547691x.2016.1173135] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Irritant contact dermatitis is the most common work-related skin disease, especially affecting workers in "wet-work" occupations. This study was conducted to investigate the association between single nucleotide polymorphisms (SNPs) within the major histocompatibility complex (MHC) and skin irritant response in a group of healthcare workers. 585 volunteer healthcare workers were genotyped for MHC SNPs and patch tested with three different irritants: sodium lauryl sulfate (SLS), sodium hydroxide (NaOH) and benzalkonium chloride (BKC). Genotyping was performed using Illumina Goldengate MHC panels. A number of SNPs within the MHC Class I (OR2B3, TRIM31, TRIM10, TRIM40 and IER3), Class II (HLA-DPA1, HLA-DPB1) and Class III (C2) genes were associated (p < 0.001) with skin response to tested irritants in different genetic models. Linkage disequilibrium patterns and functional annotations identified two SNPs in the TRIM40 (rs1573298) and HLA-DPB1 (rs9277554) genes, with a potential impact on gene regulation. In addition, SNPs in PSMB9 (rs10046277 and ITPR3 (rs499384) were associated with hand dermatitis. The results are of interest as they demonstrate that genetic variations in inflammation-related genes within the MHC can influence chemical-induced skin irritation and may explain the connection between inflamed skin and propensity to subsequent allergic contact sensitization.
Collapse
Affiliation(s)
- Berran Yucesoy
- a Health Effects Laboratory Division , CDC/NIOSH , Morgantown , WV , USA
| | - Yerkebulan Talzhanov
- b Department of Human Genetics, Graduate School of Public Health , University of Pittsburgh , Pittsburgh , PA , USA
| | - M Michael Barmada
- b Department of Human Genetics, Graduate School of Public Health , University of Pittsburgh , Pittsburgh , PA , USA
| | | | - Michael L Kashon
- a Health Effects Laboratory Division , CDC/NIOSH , Morgantown , WV , USA
| | - Elma Baron
- d University Hospitals Case Medical Center, Case Western Reserve University , Cleveland , OH , USA
| | - Nevin W Wilson
- e Department of Pediatrics, School of Medicine , University of Nevada , Reno , NV , USA
| | - Bonnie Frye
- a Health Effects Laboratory Division , CDC/NIOSH , Morgantown , WV , USA
| | - Wei Wang
- a Health Effects Laboratory Division , CDC/NIOSH , Morgantown , WV , USA
| | - Kara Fluharty
- a Health Effects Laboratory Division , CDC/NIOSH , Morgantown , WV , USA
| | - Rola Gharib
- f Department of Dermatology, School of Medicine , West Virginia University , Morgantown , WV , USA
| | - Jean Meade
- g Office of Director, CDC/NIOSH , Morgantown , WV , USA
| | - Dori Germolec
- h Toxicology Branch, DNTP/NIEHS, Research Triangle Park , NC , USA
| | - Michael I Luster
- i School of Public Health, West Virginia University , Morgantown , WV , USA
| | - Susan Nedorost
- d University Hospitals Case Medical Center, Case Western Reserve University , Cleveland , OH , USA
| |
Collapse
|
179
|
Abstract
OBJECTIVE The aim of this study was to investigate the association of single nucleotide polymorphisms (SNPs) within genes involved in inflammation, skin barrier integrity, signaling/pattern recognition, and antioxidant defense with irritant susceptibility in a group of health care workers. METHODS The 536 volunteer subjects were genotyped for selected SNPs and patch tested with three model irritants: sodium lauryl sulfate (SLS), sodium hydroxide (NaOH), and benzalkonium chloride (BKC). Genotyping was performed on genomic DNA using Illumina Goldengate custom panels. RESULTS The ACACB (rs2268387, rs16934132, rs2284685), NTRK2 (rs10868231), NTRK3 (rs1347424), IL22 (rs1179251), PLAU (rs2227564), EGFR (rs6593202), and FGF2 (rs308439) SNPs showed an association with skin response to tested irritants in different genetic models (all at P < 0.001). Functional annotations identified two SNPs in PLAU (rs2227564) and ACACB (rs2284685) genes with a potential impact on gene regulation. In addition, EGF (rs10029654), EGFR (rs12718939), CXCL12 (rs197452), and VCAM1 (rs3917018) genes showed an association with hand dermatitis (P < 0.005). CONCLUSIONS The results demonstrate that genetic variations in genes related to inflammation and skin homeostasis can influence responses to irritants and may explain inter-individual variation in the development of subsequent contact dermatitis.
Collapse
|
180
|
Kim MJ, Yu CY, Theusch E, Naidoo D, Stevens K, Kuang YL, Schuetz E, Chaudhry AS, Medina MW. SUGP1 is a novel regulator of cholesterol metabolism. Hum Mol Genet 2016; 25:3106-3116. [PMID: 27206982 PMCID: PMC5181593 DOI: 10.1093/hmg/ddw151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/05/2016] [Accepted: 05/13/2016] [Indexed: 12/19/2022] Open
Abstract
A large haplotype on chromosome 19p13.11 tagged by rs10401969 in intron 8 of SURP and G patch domain containing 1 (SUGP1) is associated with coronary artery disease (CAD), plasma LDL cholesterol levels, and other energy metabolism phenotypes. Recent studies have suggested that TM6SF2 is the causal gene within the locus, but we postulated that this locus could harbor additional CAD risk genes, including the putative splicing factor SUGP1. Indeed, we found that rs10401969 regulates SUGP1 exon 8 skipping, causing non-sense-mediated mRNA decay. Hepatic Sugp1 overexpression in CD1 male mice increased plasma cholesterol levels 20–50%. In human hepatoma cell lines, SUGP1 knockdown stimulated 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) alternative splicing and decreased HMGCR transcript stability, thus reducing cholesterol synthesis and increasing LDL uptake. Our results strongly support a role for SUGP1 as a novel regulator of cholesterol metabolism and suggest that it contributes to the relationship between rs10401969 and plasma cholesterol.
Collapse
Affiliation(s)
- Mee J Kim
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Chi-Yi Yu
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Elizabeth Theusch
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Devesh Naidoo
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Kristen Stevens
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Yu-Lin Kuang
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Erin Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amarjit S Chaudhry
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marisa W Medina
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| |
Collapse
|
181
|
Multivariate eQTL mapping uncovers functional variation on the X-chromosome associated with complex disease traits. Hum Genet 2016; 135:827-39. [PMID: 27155841 DOI: 10.1007/s00439-016-1674-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/24/2016] [Indexed: 10/21/2022]
Abstract
Very few studies have investigated the associations between genetic polymorphisms and gene expression on the X-chromosome. This is a major bottleneck when conducting functional follow-up studies of trait-associated variants, as those identified in genome-wide association studies (GWAS). We used a multivariate approach to test the association between individual single nucleotide polymorphisms (SNPs) and exon expression levels measured in 356 Epstein-Barr virus-transformed lymphoblastoid cell lines (LCLs) from the Geuvadis RNA sequencing project to identify SNPs associated with variation in gene expression on the X-chromosome, which we refer to as eSNPs. At an FDR of 5 %, we discovered 548 independent [linkage disequilibrium (LD) r (2) < 0.1] eSNPs on the X-chromosome. Of these, 35 were in LD (r (2) > 0.8) with previously published disease- or trait-associated variants identified through GWAS. One of the strongest eSNPs identified was rs35975601, which was associated with F8A1 expression (p value = 3 × 10(-20)) and was in LD with a type 1 diabetes risk variant. Additionally, we identified a number of genes for which eSNPs were in LD with multiple diseases or traits, including DNASE1L1 which was mapped to bilirubin levels, type 1 diabetes and schizophrenia. Our results also indicate that multivariate exon-level analysis provides a more powerful approach than univariate gene-level analysis, particularly when SNPs influence the expression of different exons with different magnitude and/or direction of effect. The associations identified in our study may provide new insights into the molecular process by which gene expression may contribute to trait variation or disease risk in humans.
Collapse
|
182
|
Weirather JL, Duggal P, Nascimento EL, Monteiro GR, Martins DR, Lacerda HG, Fakiola M, Blackwell JM, Jeronimo SMB, Wilson ME. Fine mapping under linkage peaks for symptomatic or asymptomatic outcomes of Leishmania infantum infection in Brazil. INFECTION GENETICS AND EVOLUTION 2016; 43:1-5. [PMID: 27155051 DOI: 10.1016/j.meegid.2016.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/08/2016] [Accepted: 05/04/2016] [Indexed: 01/08/2023]
Abstract
Infection with the protozoan Leishmania infantum can lead to asymptomatic infection and protective immunity, or to the progressive and potentially fatal disease visceral leishmaniasis (VL). Published studies show host genetic background determines in part whether infected individuals will develop a symptomatic or asymptomatic outcome. The purpose of the current study was to fine map chromosome regions previously linked with risk for symptomatic (chromosome 9) or asymptomatic (chromosomes 15 and 19) manifestations of L. infantum infection. We conducted a family-based genetic study of VL and asymptomatic infection (detected by a DTH skin test) with a final post quality control sample of 961 individuals with full genotype and phenotype information from highly endemic neighborhoods of northeast Brazil. A total of 5485 SNPs under the linkage peaks on chromosomes 9, 15 and 19 were genotyped. No strong SNP associations were observed for the DTH phenotype. The most significant associations with the VL phenotype were with SNP rs1470217 (p=5.9e-05; pcorrected=0.057) on chromosome 9, and with SNP rs8107014 (p=1.4e-05; pcorrected=0.013) on chromosome 19. SNP rs1470217 is situated in a 180kb intergenic region between TMEM215 (Transmembrane protein 215) and APTX (Aprataxin). SNP rs8107014 lies in the intron between exons 26 and 27 of a 34 exon transcript (ENST00000204005) of LTBP4, (Latent transforming growth factor-beta-binding protein 4a). The latter supports growing evidence that the transforming growth factor-beta pathway is important in the immunopathogenesis of VL.
Collapse
Affiliation(s)
- Jason L Weirather
- Interdisciplinary Program in Genetics, University of Iowa, Iowa City, IA, USA.
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Eliana L Nascimento
- Department of Infectious Diseases, Federal University of Rio Grande do Norte, Natal, RN, Brazil; Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Gloria R Monteiro
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Daniella R Martins
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Henio G Lacerda
- Department of Infectious Diseases, Federal University of Rio Grande do Norte, Natal, RN, Brazil; Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Michaela Fakiola
- Cambridge Institute for Medical Research, University of Cambridge, UK.
| | - Jenefer M Blackwell
- Cambridge Institute for Medical Research, University of Cambridge, UK; Telethon Kids Institute, The University of Western Australia, Perth, Australia.
| | - Selma M B Jeronimo
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, RN, Brazil; Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil; Institute of Science and Technology of Tropical Diseases, Brazil.
| | - Mary E Wilson
- Interdisciplinary Program in Genetics, University of Iowa, Iowa City, IA, USA; Department of Internal Medicine, University of Iowa, Iowa City, IA, USA; Department of Microbiology, University of Iowa, Iowa City, IA, USA; Iowa City Veterans' Affairs Medical Center, Iowa City, IA, USA.
| |
Collapse
|
183
|
Identification of Susceptibility Genes of Adult Asthma in French Canadian Women. Can Respir J 2016; 2016:3564341. [PMID: 27445529 PMCID: PMC4904514 DOI: 10.1155/2016/3564341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/05/2015] [Indexed: 11/18/2022] Open
Abstract
Susceptibility genes of asthma may be more successfully identified by studying subgroups of phenotypically similar asthma patients. This study aims to identify single nucleotide polymorphisms (SNPs) associated with asthma in French Canadian adult women. A pooling-based genome-wide association study was performed in 240 allergic asthmatic and 120 allergic nonasthmatic women. The top associated SNPs were selected for individual genotyping in an extended cohort of 349 asthmatic and 261 nonasthmatic women. The functional impact of asthma-associated SNPs was investigated in a lung expression quantitative trait loci (eQTL) mapping study (n = 1035). Twenty-one of the 38 SNPs tested by individual genotyping showed P values lower than 0.05 for association with asthma. Cis-eQTL analyses supported the functional contribution of rs17801353 associated with C3AR1 (P = 7.90E - 10). The asthma risk allele for rs17801353 is associated with higher mRNA expression levels of C3AR1 in lung tissue. In silico functional characterization of the asthma-associated SNPs also supported the contribution of C3AR1 and additional genes including SYNE1, LINGO2, and IFNG-AS1. This pooling-based GWAS in French Canadian adult women followed by lung eQTL mapping suggested C3AR1 as a functional locus associated with asthma. Additional susceptibility genes were suggested in this homogenous subgroup of asthma patients.
Collapse
|
184
|
Reschen ME, Lin D, Chalisey A, Soilleux EJ, O'Callaghan CA. Genetic and environmental risk factors for atherosclerosis regulate transcription of phosphatase and actin regulating gene PHACTR1. Atherosclerosis 2016; 250:95-105. [PMID: 27187934 PMCID: PMC4917897 DOI: 10.1016/j.atherosclerosis.2016.04.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/20/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Background and aims Coronary artery disease (CAD) risk is associated with non-coding genetic variants at the phosphatase and actin regulating protein 1(PHACTR1) gene locus. The PHACTR1 gene encodes an actin-binding protein with phosphatase regulating activity. The mechanism whereby PHACTR1 influences CAD risk is unknown. We hypothesized that PHACTR1 would be expressed in human cell types relevant to CAD and regulated by atherogenic or genetic factors. Methods and results Using immunohistochemistry, we demonstrate that PHACTR1 protein is expressed strongly in human atherosclerotic plaque macrophages, lipid-laden foam cells, adventitial lymphocytes and endothelial cells. Using a combination of genomic analysis and molecular techniques, we demonstrate that PHACTR1 is expressed as multiple previously uncharacterized transcripts in macrophages, foam cells, lymphocytes and endothelial cells. Immunoblotting confirmed a total absence of PHACTR1 in vascular smooth muscle cells. Real-time quantitative PCR showed that PHACTR1 is regulated by atherogenic and inflammatory stimuli. In aortic endothelial cells, oxLDL and TNF-alpha both upregulated an intermediate length transcript. A short transcript expressed only in immune cells was upregulated in macrophages by oxidized low-density lipoprotein, and oxidized phospholipids but suppressed by lipopolysaccharide or TNF-alpha. In primary human macrophages, we identified a novel expression quantitative trait locus (eQTL) specific for this short transcript, whereby the risk allele at CAD risk SNP rs9349379 is associated with reduced PHACTR1 expression, similar to the effect of an inflammatory stimulus. Conclusions Our data demonstrate that PHACTR1 is a key atherosclerosis candidate gene since it is regulated by atherogenic stimuli in macrophages and endothelial cells and we identify an effect of the genetic risk variant on PHACTR1 expression in macrophages that is similar to that of an inflammatory stimulus. PHACTR1 is expressed as two transcripts in both immune and endothelial cells in human atherosclerotic plaque. Oxidized-LDL upregulates a short PHACTR1 transcript, but suppresses an intermediate length transcript in macrophages. Lipopolysaccharide and TNF-alpha cause the opposite effect with strong suppression of the short transcript in macrophages. The coronary artery disease risk SNP, rs9349379, is associated with expression of the short transcript in macrophages. The effect of the coronary artery disease risk allele on PHACTR1 mirrors that of inflammatory stimuli.
Collapse
Affiliation(s)
- Michael E Reschen
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Da Lin
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Anil Chalisey
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom
| | - Elizabeth J Soilleux
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford and Department of Cellular Pathology, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Christopher A O'Callaghan
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, United Kingdom.
| |
Collapse
|
185
|
Peterson CB, Service SK, Jasinska AJ, Gao F, Zelaya I, Teshiba TM, Bearden CE, Cantor RM, Reus VI, Macaya G, López-Jaramillo C, Bogomolov M, Benjamini Y, Eskin E, Coppola G, Freimer NB, Sabatti C. Characterization of Expression Quantitative Trait Loci in Pedigrees from Colombia and Costa Rica Ascertained for Bipolar Disorder. PLoS Genet 2016; 12:e1006046. [PMID: 27176483 PMCID: PMC4866754 DOI: 10.1371/journal.pgen.1006046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/20/2016] [Indexed: 01/22/2023] Open
Abstract
The observation that variants regulating gene expression (expression quantitative trait loci, eQTL) are at a high frequency among SNPs associated with complex traits has made the genome-wide characterization of gene expression an important tool in genetic mapping studies of such traits. As part of a study to identify genetic loci contributing to bipolar disorder and other quantitative traits in members of 26 pedigrees from Costa Rica and Colombia, we measured gene expression in lymphoblastoid cell lines derived from 786 pedigree members. The study design enabled us to comprehensively reconstruct the genetic regulatory network in these families, provide estimates of heritability, identify eQTL, evaluate missing heritability for the eQTL, and quantify the number of different alleles contributing to any given locus. In the eQTL analysis, we utilize a recently proposed hierarchical multiple testing strategy which controls error rates regarding the discovery of functional variants. Our results elucidate the heritability and regulation of gene expression in this unique Latin American study population and identify a set of regulatory SNPs which may be relevant in future investigations of complex disease in this population. Since our subjects belong to extended families, we are able to compare traditional kinship-based estimates with those from more recent methods that depend only on genotype information.
Collapse
Affiliation(s)
- Christine B. Peterson
- Department of Health Research and Policy, Stanford University, Stanford, California, United States of America
| | - Susan K. Service
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Anna J. Jasinska
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Fuying Gao
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
| | - Ivette Zelaya
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
| | - Terri M. Teshiba
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rita M. Cantor
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Victor I. Reus
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
| | - Gabriel Macaya
- Cell and Molecular Biology Research Center, Universidad de Costa Rica, San Pedro de Montes de Oca, San José, Costa Rica
| | - Carlos López-Jaramillo
- Grupo de Investigación en Psiquiatría (Research Group in Psychiatry (GIPSI)), Departamento de Psiquiatría, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia; Mood Disorders Program, Hospital San Vicente Fundacion, Medellín, Colombia
| | - Marina Bogomolov
- Faculty of Industrial Engineering and Management, Technion, Haifa, Israel
| | - Yoav Benjamini
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Eleazar Eskin
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Computer Science, University of California Los Angeles, Los Angeles, California, United States of America
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Nelson B. Freimer
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, United States of America
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Chiara Sabatti
- Department of Biomedical Data Science and Department of Statistics, Stanford University, Stanford, California, United States of America
| |
Collapse
|
186
|
Müller-Calleja N, Rossmann H, Müller C, Wild P, Blankenberg S, Pfeiffer N, Binder H, Beutel ME, Manukyan D, Zeller T, Lackner KJ. Antiphospholipid antibodies in a large population-based cohort: genome-wide associations and effects on monocyte gene expression. Thromb Haemost 2016; 116:115-23. [PMID: 27098658 DOI: 10.1160/th15-12-0947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/15/2016] [Indexed: 11/05/2022]
Abstract
The antiphospholipid syndrome (APS) is characterised by venous and/or arterial thrombosis and pregnancy morbidity in women combined with the persistent presence of antiphospholipid antibodies (aPL). We aimed to identify genetic factors associated with the presence of aPL in a population based cohort. Furthermore, we wanted to clarify if the presence of aPL affects gene expression in circulating monocytes. Titres of IgG and IgM against cardiolipin, β2glycoprotein 1 (anti-β2GPI), and IgG against domain 1 of β2GPI (anti-domain 1) were determined in approx. 5,000 individuals from the Gutenberg Health Study (GHS) a population based cohort of German descent. Genotyping was conducted on Affymetrix Genome-Wide Human SNP 6.0 arrays. Monocyte gene expression was determined in a subgroup of 1,279 individuals by using the Illumina HT-12 v3 BeadChip. Gene expression data were confirmed in vitro and ex vivo by qRT-PCR. Genome wide analysis revealed significant associations of anti-β2GPI IgG and APOH on chromosome 17, which had been previously identified by candidate gene approaches, and of anti-domain1 and MACROD2 on chromosome 20 which has been listed in a previous GWAS as a suggestive locus associated with the occurrence of anti-β2GPI antibodies. Expression analysis confirmed increased expression of TNFα in monocytes and identified and confirmed neuron navigator 3 (NAV3) as a novel gene induced by aPL. In conclusion, MACROD2 represents a novel genetic locus associated with aPL. Furthermore, we show that aPL induce the expression of NAV3 in monocytes and endothelial cells. This will stimulate further research into the role of these genes in the APS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karl J Lackner
- Dr. Karl J. Lackner, Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany, Tel.: +49 6131 177190, E-mail:
| |
Collapse
|
187
|
Peterson CB, Bogomolov M, Benjamini Y, Sabatti C. TreeQTL: hierarchical error control for eQTL findings. ACTA ACUST UNITED AC 2016; 32:2556-8. [PMID: 27153635 DOI: 10.1093/bioinformatics/btw198] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 04/10/2016] [Indexed: 12/24/2022]
Abstract
UNLABELLED : Commonly used multiplicity adjustments fail to control the error rate for reported findings in many expression quantitative trait loci (eQTL) studies. TreeQTL implements a hierarchical multiple testing procedure which allows control of appropriate error rates defined relative to a grouping of the eQTL hypotheses. AVAILABILITY AND IMPLEMENTATION The R package TreeQTL is available for download at http://bioinformatics.org/treeqtl CONTACT sabatti@stanford.edu SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- C B Peterson
- Department of Health Research and Policy, Stanford University, Stanford, CA 94305, USA
| | - M Bogomolov
- Faculty of Industrial Engineering and Management, Technion, Haifa 32000, Israel
| | - Y Benjamini
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv 6997801, Israel
| | - C Sabatti
- Departments of Biomedical Data Science and Statistics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
188
|
Rizzu P, Blauwendraat C, Heetveld S, Lynes EM, Castillo-Lizardo M, Dhingra A, Pyz E, Hobert M, Synofzik M, Simón-Sánchez J, Francescatto M, Heutink P. C9orf72 is differentially expressed in the central nervous system and myeloid cells and consistently reduced in C9orf72, MAPT and GRN mutation carriers. Acta Neuropathol Commun 2016; 4:37. [PMID: 27079381 PMCID: PMC4832459 DOI: 10.1186/s40478-016-0306-7] [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: 03/25/2016] [Accepted: 03/25/2016] [Indexed: 12/12/2022] Open
Abstract
A non-coding hexanucleotide repeat expansion (HRE) in C9orf72 is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) acting through a loss of function mechanism due to haploinsufficiency of C9orf72 or a gain of function mediated by aggregates of bidirectionally transcribed HRE-RNAs translated into di-peptide repeat (DPR) proteins. To fully understand regulation of C9orf72 expression we surveyed the C9orf72 locus using Cap Analysis of Gene Expression sequence data (CAGEseq). We observed C9orf72 was generally lowly expressed with the exception of a subset of myeloid cells, particularly CD14+ monocytes that showed up to seven fold higher expression as compared to central nervous system (CNS) and other tissues. The expression profile at the C9orf72 locus showed a complex architecture with differential expression of the transcription start sites (TSSs) for the annotated C9orf72 transcripts between myeloid and CNS tissues suggesting cell and/or tissue specific functions. We further detected novel TSSs in both the sense and antisense strand at the C9orf72 locus and confirmed their existence in brain tissues and CD14+ monocytes. Interestingly, our experiments showed a consistent decrease of C9orf72 coding transcripts not only in brain tissue and monocytes from C9orf72-HRE patients, but also in brains from MAPT and GRN mutation carriers together with an increase in antisense transcripts suggesting these could play a role in regulation of C9orf72. We found that the non-HRE related expression changes cannot be explained by promoter methylation but by the presence of the C9orf72-HRE risk haplotype and unknown functional interactions between C9orf72, MAPT and GRN.
Collapse
|
189
|
Johansson Å, Eriksson N, Lindholm D, Varenhorst C, James S, Syvänen AC, Axelsson T, Siegbahn A, Barratt BJ, Becker RC, Himmelmann A, Katus HA, Steg PG, Storey RF, Wallentin L. Genome-wide association and Mendelian randomization study of NT-proBNP in patients with acute coronary syndrome. Hum Mol Genet 2016; 25:1447-56. [PMID: 26908625 DOI: 10.1093/hmg/ddw012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/14/2016] [Indexed: 01/28/2023] Open
Abstract
N-terminal pro-B-type natriuretic peptide (NT-proBNP) is a strong predictor of mortality in coronary artery disease and is widely employed as a prognostic biomarker. However, a causal relationship between NT-proBNP and clinical endpoints has not been established. We have performed a genome-wide association and Mendelian randomization study of NT-proBNP. We used a discovery set of 3740 patients from the PLATelet inhibition and patient Outcomes (PLATO) trial, which enrolled 18 624 patients with acute coronary syndrome (ACS). A further set of 5492 patients, from the same trial, was used for replication. Genetic variants at two novel loci (SLC39A8 and POC1B/GALNT4) were associated with NT-proBNP levels and replicated together with the previously known NPPB locus. The most significant SNP (rs198389, pooled P = 1.07 × 10(-15)) in NPPB interrupts an E-box consensus motif in the gene promoter. The association in SLC39A8 is driven by a deleterious variant (rs13107325, pooled P = 5.99 × 10(-10)), whereas the most significant SNP in POC1B/GALNT4 (rs11105306, pooled P = 1.02 × 10(-16)) is intronic. The SLC39A8 SNP was associated with higher risk of cardiovascular (CV) death (HR = 1.39, 95% CI: 1.08-1.79, P = 0.0095), but the other loci were not associated with clinical endpoints. We have identified two novel loci to be associated with NT-proBNP in patients with ACS. Only the SLC39A8 variant, but not the NPPB variant, was associated with a clinical endpoint. Due to pleotropic effects of SLC39A8, these results do not suggest that NT-proBNP levels have a direct effect on mortality in ACS patients. PLATO Clinical Trial Registration: www.clinicaltrials.gov; NCT00391872.
Collapse
Affiliation(s)
- Åsa Johansson
- Uppsala Clinical Research Center and Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 752 37, Sweden,
| | | | - Daniel Lindholm
- Uppsala Clinical Research Center and Department of Medical Sciences, Cardiology
| | | | - Stefan James
- Uppsala Clinical Research Center and Department of Medical Sciences, Cardiology
| | | | - Tomas Axelsson
- Department of Medical Sciences, Molecular Medicine, Science for Life Laboratory and
| | - Agneta Siegbahn
- Uppsala Clinical Research Center and Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala 751 85, Sweden
| | | | - Richard C Becker
- Division of Cardiovascular Health and Disease, Heart, Lung and Vascular Institute, Academic Health Center, Cincinnati, OH 45267-0542, USA
| | | | - Hugo A Katus
- Medizinishe Klinik, Universitätsklinikum Heidelberg, Heidelberg 69120, Germany
| | - Philippe Gabriel Steg
- INSERM-Unité 1148, Paris 75019, France, Assistance Publique-Hôpitaux de Paris, Département Hospitalo-Universitaire FIRE, Hôpital Bichat, Paris 75018, France, Université Paris-Diderot, Sorbonne-Paris Cité, Paris 75013, France, NHLI Imperial College, ICMS, Royal Brompton Hospital, London SW3 6NP, UK and
| | - Robert F Storey
- Department of Cardiovascular Science, University of Sheffield, Sheffield S10 2RX, UK
| | - Lars Wallentin
- Uppsala Clinical Research Center and Department of Medical Sciences, Cardiology
| |
Collapse
|
190
|
Stracquadanio G, Wang X, Wallace M, Grawenda AM, Zhang P, Hewitt J, Zeron-Medina J, Castro-Giner F, Tomlinson IP, Goding CR, Cygan KJ, Fairbrother WG, Thomas LF, Sætrom P, Gemignani F, Landi S, Schuster-Boeckler B, Bell DA, Bond GL. The importance of p53 pathway genetics in inherited and somatic cancer genomes. Nat Rev Cancer 2016; 16:251-65. [PMID: 27009395 PMCID: PMC6854702 DOI: 10.1038/nrc.2016.15] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decades of research have shown that mutations in the p53 stress response pathway affect the incidence of diverse cancers more than mutations in other pathways. However, most evidence is limited to somatic mutations and rare inherited mutations. Using newly abundant genomic data, we demonstrate that commonly inherited genetic variants in the p53 pathway also affect the incidence of a broad range of cancers more than variants in other pathways. The cancer-associated single nucleotide polymorphisms (SNPs) of the p53 pathway have strikingly similar genetic characteristics to well-studied p53 pathway cancer-causing somatic mutations. Our results enable insights into p53-mediated tumour suppression in humans and into p53 pathway-based cancer surveillance and treatment strategies.
Collapse
Affiliation(s)
- Giovanni Stracquadanio
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Xuting Wang
- Environmental Genomics Group, Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Marsha Wallace
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Anna M. Grawenda
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Ping Zhang
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Juliet Hewitt
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Jorge Zeron-Medina
- Vall d’Hebron University Hospital, Oncology Department, Passeig de la Vall D’Hebron 119, 08035 Barcelona, Spain
| | - Francesc Castro-Giner
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Ian P. Tomlinson
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Colin R. Goding
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Kamil J. Cygan
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - William G. Fairbrother
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Laurent F. Thomas
- Department of Cancer Research and Molecular Medicine, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
| | - Pål Sætrom
- Department of Computer and Information Science, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian, University of Science and Technology, NO-7491 Trondheim, Norway
| | - Frederica Gemignani
- Genetics- Department of Biology, University of Pisa, Via Derna, 1, 56126 Pisa - Italy
| | - Stefano Landi
- Genetics- Department of Biology, University of Pisa, Via Derna, 1, 56126 Pisa - Italy
| | - Benjamin Schuster-Boeckler
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Douglas A. Bell
- Environmental Genomics Group, Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
- Corresponding authors: . The Ludwig Institute for Cancer Research, The Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, The United Kingdom. . Environmental Genomics Group, Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, MD C3-03, NIEHS, PO Box 12233, Research Triangle Park, NC 27709, The United States of America
| | - Gareth L. Bond
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
- Corresponding authors: . The Ludwig Institute for Cancer Research, The Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, The United Kingdom. . Environmental Genomics Group, Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, MD C3-03, NIEHS, PO Box 12233, Research Triangle Park, NC 27709, The United States of America
| |
Collapse
|
191
|
Engelken J, Espadas G, Mancuso FM, Bonet N, Scherr AL, Jímenez-Álvarez V, Codina-Solà M, Medina-Stacey D, Spataro N, Stoneking M, Calafell F, Sabidó E, Bosch E. Signatures of Evolutionary Adaptation in Quantitative Trait Loci Influencing Trace Element Homeostasis in Liver. Mol Biol Evol 2016; 33:738-54. [PMID: 26582562 PMCID: PMC4760079 DOI: 10.1093/molbev/msv267] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Essential trace elements possess vital functions at molecular, cellular, and physiological levels in health and disease, and they are tightly regulated in the human body. In order to assess variability and potential adaptive evolution of trace element homeostasis, we quantified 18 trace elements in 150 liver samples, together with the expression levels of 90 genes and abundances of 40 proteins involved in their homeostasis. Additionally, we genotyped 169 single nucleotide polymorphism (SNPs) in the same sample set. We detected significant associations for 8 protein quantitative trait loci (pQTL), 10 expression quantitative trait loci (eQTLs), and 15 micronutrient quantitative trait loci (nutriQTL). Six of these exceeded the false discovery rate cutoff and were related to essential trace elements: 1) one pQTL for GPX2 (rs10133290); 2) two previously described eQTLs for HFE (rs12346) and SELO (rs4838862) expression; and 3) three nutriQTLs: The pathogenic C282Y mutation at HFE affecting iron (rs1800562), and two SNPs within several clustered metallothionein genes determining selenium concentration (rs1811322 and rs904773). Within the complete set of significant QTLs (which involved 30 SNPs and 20 gene regions), we identified 12 SNPs with extreme patterns of population differentiation (FST values in the top 5% percentile in at least one HapMap population pair) and significant evidence for selective sweeps involving QTLs at GPX1, SELENBP1, GPX3, SLC30A9, and SLC39A8. Overall, this detailed study of various molecular phenotypes illustrates the role of regulatory variants in explaining differences in trace element homeostasis among populations and in the human adaptive response to environmental pressures related to micronutrients.
Collapse
Affiliation(s)
- Johannes Engelken
- †These authors contributed equally to this work. ‡Deceased October 23, 2015. Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain Department of Evolutionary Genetics, Max-Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Guadalupe Espadas
- †These authors contributed equally to this work. Proteomics Unit, Center of Genomics Regulation, Barcelona, Spain Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Francesco M Mancuso
- Proteomics Unit, Center of Genomics Regulation, Barcelona, Spain Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nuria Bonet
- Genomics Core Facility, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Anna-Lena Scherr
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Victoria Jímenez-Álvarez
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Codina-Solà
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Daniel Medina-Stacey
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nino Spataro
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mark Stoneking
- Department of Evolutionary Genetics, Max-Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Francesc Calafell
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Center of Genomics Regulation, Barcelona, Spain Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Elena Bosch
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
192
|
Chen S, Huang J, Zhao Q, Chen J, Jaquish CE, He J, Lu X, Yang X, Gu CC, Hixson JE, Liu F, Rice TK, Cao J, Chen J, Gu D. Associations Between Genetic Variants of the Natriuretic Peptide System and Blood Pressure Response to Dietary Sodium Intervention: The GenSalt Study. Am J Hypertens 2016. [PMID: 26224401 DOI: 10.1093/ajh/hpv129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The aim of this study was to comprehensively test the association of genetic variants in the natriuretic peptide (NP) system with blood pressure (BP) response to dietary sodium intervention in a Chinese population. METHODS We conducted a 7-day low-sodium intervention followed by a 7-day high-sodium intervention among 1,906 participants in rural China. BP measurements were obtained at baseline and each dietary intervention using a random-zero sphygmomanometer. Linear mixed-effect models were used to assess the associations of 48 single-nucleotide polymorphisms (SNPs) in 6 genes of NP system with BP response to dietary sodium intervention. RESULTS SNP rs5063 in the NPPA gene and SNP rs2077386 in the NPPC gene exhibited significant associations with BP response to low-sodium dietary intervention under recessive genetic model. For rs5063, absolute mean arterial pressure responses (95% confidence interval) to the low-sodium intervention were 1.31 (-1.08, 3.70) mm Hg for TT genotype and -3.74 (-4.01, -3.46) mm Hg for CC or TC genotype, respectively (P = 4.1 × 10(-5)). Individuals with at least one copy of the C allele of rs2077386 had significantly reduction in systolic BP during the low-sodium intervention compared to those with genotype GG with responses of -5.48 (-5.83, -5.14) vs. -2.76 (-3.52, -2.00) mm Hg, respectively (P = 1.9 × 10(-13)). CONCLUSIONS These novel findings suggested that genetic variants of NP system may contribute to the variation of BP response to sodium intervention in Chinese population. Certainly, replication of these results in other populations and further functional studies are warranted to clarify their role in the regulation of BP and hypertension.
Collapse
Affiliation(s)
- Shufeng Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China;
| | - Jianfeng Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Zhao
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jing Chen
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Cashell E Jaquish
- National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueli Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Charles C Gu
- School of Medicine, Washington University, St. Louis, Missouri, USA
| | - James E Hixson
- School of Public Health, University of Texas, Houston, Texas, USA
| | - Fangchao Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Treva K Rice
- School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Jie Cao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jichun Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongfeng Gu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
193
|
Karch CM, Ezerskiy LA, Bertelsen S, Goate AM. Alzheimer's Disease Risk Polymorphisms Regulate Gene Expression in the ZCWPW1 and the CELF1 Loci. PLoS One 2016; 11:e0148717. [PMID: 26919393 PMCID: PMC4769299 DOI: 10.1371/journal.pone.0148717] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022] Open
Abstract
Late onset Alzheimer’s disease (LOAD) is a genetically complex and clinically heterogeneous disease. Recent large-scale genome wide association studies (GWAS) have identified more than twenty loci that modify risk for AD. Despite the identification of these loci, little progress has been made in identifying the functional variants that explain the association with AD risk. Thus, we sought to determine whether the novel LOAD GWAS single nucleotide polymorphisms (SNPs) alter expression of LOAD GWAS genes and whether expression of these genes is altered in AD brains. The majority of LOAD GWAS SNPs occur in gene dense regions under large linkage disequilibrium (LD) blocks, making it unclear which gene(s) are modified by the SNP. Thus, we tested for brain expression quantitative trait loci (eQTLs) between LOAD GWAS SNPs and SNPs in high LD with the LOAD GWAS SNPs in all of the genes within the GWAS loci. We found a significant eQTL between rs1476679 and PILRB and GATS, which occurs within the ZCWPW1 locus. PILRB and GATS expression levels, within the ZCWPW1 locus, were also associated with AD status. Rs7120548 was associated with MTCH2 expression, which occurs within the CELF1 locus. Additionally, expression of several genes within the CELF1 locus, including MTCH2, were highly correlated with one another and were associated with AD status. We further demonstrate that PILRB, as well as other genes within the GWAS loci, are most highly expressed in microglia. These findings together with the function of PILRB as a DAP12 receptor supports the critical role of microglia and neuroinflammation in AD risk.
Collapse
Affiliation(s)
- Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (CMK); (AMG)
| | - Lubov A. Ezerskiy
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sarah Bertelsen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States of America
| | | | - Alison M. Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States of America
- * E-mail: (CMK); (AMG)
| |
Collapse
|
194
|
A variant at 9p21.3 functionally implicates CDKN2B in paediatric B-cell precursor acute lymphoblastic leukaemia aetiology. Nat Commun 2016; 7:10635. [PMID: 26868379 PMCID: PMC4754340 DOI: 10.1038/ncomms10635] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 01/07/2016] [Indexed: 01/25/2023] Open
Abstract
Paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) is the most common cancer of childhood, yet little is known about BCP-ALL predisposition. In this study, in 2,187 cases of European ancestry and 5,543 controls, we discover and replicate a locus indexed by rs77728904 at 9p21.3 associated with BCP-ALL susceptibility (Pcombined=3.32 × 10(-15), OR=1.72) and independent from rs3731217, the previously reported ALL-associated variant in this region. Of correlated SNPs tagged by this locus, only rs662463 is significant in African Americans, suggesting it is a plausible causative variant. Functional analysis shows that rs662463 is a cis-eQTL for CDKN2B, with the risk allele associated with lower expression, and suggests that rs662463 influences BCP-ALL risk by regulating CDKN2B expression through CEBPB signalling. Functional analysis of rs3731217 suggests it is associated with BCP-ALL by acting within a splicing regulatory element determining CDKN2A exon 3 usage (P=0.01). These findings provide new insights into the critical role of the CDKN2 locus in BCP-ALL aetiology.
Collapse
|
195
|
Ebbert MTW, Boehme KL, Wadsworth ME, Staley LA, Mukherjee S, Crane PK, Ridge PG, Kauwe JSK. Interaction between variants in CLU and MS4A4E modulates Alzheimer's disease risk. Alzheimers Dement 2016; 12:121-129. [PMID: 26449541 PMCID: PMC4744542 DOI: 10.1016/j.jalz.2015.08.163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/17/2015] [Accepted: 08/17/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Ebbert et al. reported gene-gene interactions between rs11136000-rs670139 (CLU-MS4A4E) and rs3865444-rs670139 (CD33-MS4A4E). We evaluate these interactions in the largest data set for an epistasis study. METHODS We tested interactions using 3837 cases and 4145 controls from Alzheimer's Disease Genetics Consortium using meta-analyses and permutation analyses. We repeated meta-analyses stratified by apolipoprotein E (APOE) ε4 status, estimated combined odds ratio (OR) and population attributable fraction (cPAF), and explored causal variants. RESULTS Results support the CLU-MS4A4E interaction and a dominant effect. An association between CLU-MS4A4E and APOE ε4 negative status exists. The estimated synergy factor, OR, and cPAF for rs11136000-rs670139 are 2.23, 2.45, and 8.0, respectively. We identified potential causal variants. DISCUSSION We replicated the CLU-MS4A4E interaction in a large case-control series and observed APOE ε4 and possible dominant effect. The CLU-MS4A4E OR is higher than any Alzheimer's disease locus except APOE ε4, APP, and TREM2. We estimated an 8% decrease in Alzheimer's disease incidence without CLU-MS4A4E risk alleles and identified potential causal variants.
Collapse
Affiliation(s)
- Mark T W Ebbert
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Kevin L Boehme
- Department of Biology, Brigham Young University, Provo, UT, USA
| | | | | | | | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Perry G Ridge
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
196
|
Shirpoor A, Norouzi L, Nemati S, Khadem Ansari MH. Protective effect of vitamin E against diabetes-induced oxidized LDL and aorta cell wall proliferation in rat. IRANIAN BIOMEDICAL JOURNAL 2016; 19:117-23. [PMID: 25864817 PMCID: PMC4412923 DOI: 10.6091/ibj.1449.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
UNLABELLED Hyperlipidemia and oxidized-low-density lipoproteins (Ox-LDL) are important independent cardiovascular risk factors that have been shown to stimulate vascular smooth muscle cell (VSMC) proliferation. The purpose of the present study was to investigate the effect of vitamin E on Ox-LDL, lipid profile, C-reactive protein (CRP), and VSMC proliferation of rat aorta. METHODS Male Wistar rats (n = 32) were divided into four groups namely: sham (SH), control (C), non-treated diabetic, and vitamin E-treated diabetic (VETD) groups. Ox-LDL, lipid profile, CRP and VSMC proliferation of aorta were measured after 42 days. RESULTS The results revealed that along with a significant increase in VSMC proliferation, the amount of CRP, Ox-LDL, and lipid profiles in diabetic rats. VSMC proliferation was significantly ameliorated, and elevated CRP, Ox-LDL, and lipid profiles were also restored to those of shams in VETD. CONCLUSIONS These findings strongly support the idea that diabetes induces Ox-LDL-mediated oxidative stress and VSMC proliferation in aorta of rat and imply that vitamin E has a strong protective effect as an antioxidant.
Collapse
Affiliation(s)
- Alireza Shirpoor
- Dept. of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Norouzi
- Dept. of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Samira Nemati
- Dept. of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | |
Collapse
|
197
|
Kim S, Welsh DA, Myers L, Cherry KE, Wyckoff J, Jazwinski SM. Non-coding genomic regions possessing enhancer and silencer potential are associated with healthy aging and exceptional survival. Oncotarget 2016; 6:3600-12. [PMID: 25682868 PMCID: PMC4414140 DOI: 10.18632/oncotarget.2877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/08/2014] [Indexed: 01/04/2023] Open
Abstract
We have completed a genome-wide linkage scan for healthy aging using data collected from a family study, followed by fine-mapping by association in a separate population, the first such attempt reported. The family cohort consisted of parents of age 90 or above and their children ranging in age from 50 to 80. As a quantitative measure of healthy aging, we used a frailty index, called FI34, based on 34 health and function variables. The linkage scan found a single significant linkage peak on chromosome 12. Using an independent cohort of unrelated nonagenarians, we carried out a fine-scale association mapping of the region suggestive of linkage and identified three sites associated with healthy aging. These healthy-aging sites (HASs) are located in intergenic regions at 12q13-14. HAS-1 has been previously associated with multiple diseases, and an enhancer was recently mapped and experimentally validated within the site. HAS-2 is a previously uncharacterized site possessing genomic features suggestive of enhancer activity. HAS-3 contains features associated with Polycomb repression. The HASs also contain variants associated with exceptional longevity, based on a separate analysis. Our results provide insight into functional genomic networks involving non-coding regulatory elements that are involved in healthy aging and longevity.
Collapse
Affiliation(s)
- Sangkyu Kim
- Tulane Center for Aging and Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - David A Welsh
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Leann Myers
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Katie E Cherry
- Department of Psychology, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Jennifer Wyckoff
- Tulane Center for Aging and Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - S Michal Jazwinski
- Tulane Center for Aging and Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
198
|
Gene Silencing and Haploinsufficiency of Csk Increase Blood Pressure. PLoS One 2016; 11:e0146841. [PMID: 26751575 PMCID: PMC4713444 DOI: 10.1371/journal.pone.0146841] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/22/2015] [Indexed: 01/11/2023] Open
Abstract
Objective Recent genome-wide association studies have identified 33 human genetic loci that influence blood pressure. The 15q24 locus is one such locus that has been confirmed in Asians and Europeans. There are 21 genes in the locus within a 1-Mb boundary, but a functional link of these genes to blood pressure has not been reported. We aimed to identify a causative gene for blood pressure change in the 15q24 locus. Methods and Results CSK and ULK3 were selected as candidate genes based on eQTL analysis studies that showed the association between gene transcript levels and the lead SNP (rs1378942). Injection of siRNAs for mouse homologs Csk, Ulk3, and Cyp1a2 (negative control) showed reduced target gene mRNA levels in vivo. However, Csk siRNA only increased blood pressure while Ulk3 and Cyp1a2 siRNA did not change it. Further, blood pressure in Csk+/- heterozygotes was higher than in wild-type, consistent with what we observed in Csk siRNA-injected mice. We confirmed that haploinsufficiency of Csk increased the active form of Src in Csk+/- mice aorta. We also showed that inhibition of Src by PP2, a Src inhibitor decreased high blood pressure in Csk+/- mice and the active Src in Csk+/- mice aorta and in Csk knock-down vascular smooth muscle cells, suggesting blood pressure regulation by Csk through Src. Conclusions Our study demonstrates that Csk is a causative gene in the 15q24 locus and regulates blood pressure through Src, and these findings provide a novel therapeutic target for the treatment of hypertension.
Collapse
|
199
|
Mao F, Xiao L, Li X, Liang J, Teng H, Cai W, Sun ZS. RBP-Var: a database of functional variants involved in regulation mediated by RNA-binding proteins. Nucleic Acids Res 2016; 44:D154-63. [PMID: 26635394 PMCID: PMC4702914 DOI: 10.1093/nar/gkv1308] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/01/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022] Open
Abstract
Transcription factors bind to the genome by forming specific contacts with the primary DNA sequence; however, RNA-binding proteins (RBPs) have greater scope to achieve binding specificity through the RNA secondary structure. It has been revealed that single nucleotide variants (SNVs) that alter RNA structure, also known as RiboSNitches, exhibit 3-fold greater local structure changes than replicates of the same DNA sequence, demonstrated by the fact that depletion of RiboSNitches could result in the alteration of specific RNA shapes at thousands of sites, including 3' UTRs, binding sites of microRNAs and RBPs. However, the network between SNVs and post-transcriptional regulation remains unclear. Here, we developed RBP-Var, a database freely available at http://www.rbp-var.biols.ac.cn/, which provides annotation of functional variants involved in post-transcriptional interaction and regulation. RBP-Var provides an easy-to-use web interface that allows users to rapidly find whether SNVs of interest can transform the secondary structure of RNA and identify RBPs whose binding may be subsequently disrupted. RBP-Var integrates DNA and RNA biology to understand how various genetic variants and post-transcriptional mechanisms cooperate to orchestrate gene expression. In summary, RBP-Var is useful in selecting candidate SNVs for further functional studies and exploring causal SNVs underlying human diseases.
Collapse
Affiliation(s)
- Fengbiao Mao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luoyuan Xiao
- Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China
| | - Xianfeng Li
- State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan, 410078, China
| | - Jialong Liang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huajing Teng
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wanshi Cai
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhong Sheng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
200
|
Liquet B, Bottolo L, Campanella G, Richardson S, Chadeau-Hyam M. R2GUESS: A Graphics Processing Unit-Based R Package for Bayesian Variable Selection Regression of Multivariate Responses. J Stat Softw 2016; 69. [PMID: 29568242 DOI: 10.18637/jss.v069.i02] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Technological advances in molecular biology over the past decade have given rise to high dimensional and complex datasets offering the possibility to investigate biological associations between a range of genomic features and complex phenotypes. The analysis of this novel type of data generated unprecedented computational challenges which ultimately led to the definition and implementation of computationally efficient statistical models that were able to scale to genome-wide data, including Bayesian variable selection approaches. While extensive methodological work has been carried out in this area, only few methods capable of handling hundreds of thousands of predictors were implemented and distributed. Among these we recently proposed GUESS, a computationally optimised algorithm making use of graphics processing unit capabilities, which can accommodate multiple outcomes. In this paper we propose R2GUESS, an R package wrapping the original C++ source code. In addition to providing a user-friendly interface of the original code automating its parametrisation, and data handling, R2GUESS also incorporates many features to explore the data, to extend statistical inferences from the native algorithm (e.g., effect size estimation, significance assessment), and to visualize outputs from the algorithm. We first detail the model and its parametrisation, and describe in details its optimised implementation. Based on two examples we finally illustrate its statistical performances and flexibility.
Collapse
Affiliation(s)
- Benoît Liquet
- Laboratoire de Mathématiques et de leurs Applications, Université de Pau et des Pays de l'Adour, UMR CNRS 5142, Pau, France; ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology (QUT), Brisbane, Australia
| | | | | | | | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Hospital, Norfolk Place, London, W21PG, United Kingdom
| |
Collapse
|