101
|
Marchesan JT, Jiao Y, Moss K, Divaris K, Seaman W, Webster-Cyriaque J, Zhang S, Yu N, Song C, Bencharit S, Teles R, Offenbacher S. Common Polymorphisms in IFI16 and AIM2 Genes Are Associated With Periodontal Disease. J Periodontol 2017; 88:663-672. [PMID: 28387608 DOI: 10.1902/jop.2017.160553] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND The single nucleotide polymorphism (SNP) context of a previously identified periodontitis-associated locus is investigated, and its association with microbial, biologic, and periodontal disease clinical parameters is examined. METHODS A 200-kb spanning region of 1q12 previously highlighted in a genome-wide association scan among 4,766 European American individuals (SNP rs1633266) was annotated. Two haplotype blocks were selected. Association of these polymorphisms with data on microbial plaque composition, gingival crevicular fluid (GCF)-interleukin (IL)-1β levels, and clinical parameters of periodontal disease were examined. Descriptive analysis of IFI16 and AIM2 protein expression in gingival tissues from healthy individuals (n = 2) and individuals with chronic periodontitis (n = 2) was done via immunohistochemistry. RESULTS The highlighted locus is a 100-kb region containing the interferon γ-inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) genes. Two haplotype blocks, rs6940 and rs1057028, were significantly associated with increased extent bleeding on probing and levels of microorganisms Porphyromonas gingivalis, Tannerella forsythia, and Campylobacter rectus (P ≤0.05). Haplotype block rs1057028 was also significantly associated with pathogens Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans, increased GCF-IL-1β levels, and extent of probing depth ≥4 mm (P ≤0.05). Prevalence of severe periodontitis (biofilm-gingival interface P3 classification) was positively associated with haplotype block rs1057028. Similar trends were observed for haplotype block rs1057028. IFI16 and AIM2 protein expression was observed in multiple cell types of gingival tissues, including inflammatory cells. CONCLUSION This study found IFI16 and AIM2 SNPs associated with higher levels of periodontal microorganisms and an increased percentage of periodontal disease clinical parameters, suggesting the need for functional studies and additional fine-mapping of variants in the 1q12-locus.
Collapse
Affiliation(s)
- Julie T Marchesan
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC.,General Oral Health Clinic, School of Dentistry, University of North Carolina at Chapel Hill
| | - Yizu Jiao
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kevin Moss
- General Oral Health Clinic, School of Dentistry, University of North Carolina at Chapel Hill
| | - Kimon Divaris
- Department of Pediatric Dentistry, School of Dentistry and Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - William Seaman
- UNC Lineberger, School of Medicine, University of North Carolina at Chapel Hill
| | - Jennifer Webster-Cyriaque
- UNC Lineberger, School of Medicine, University of North Carolina at Chapel Hill.,Department of Dental Ecology, School of Dentistry, University of North Carolina at Chapel Hill
| | - Shaoping Zhang
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC.,General Oral Health Clinic, School of Dentistry, University of North Carolina at Chapel Hill
| | - Ning Yu
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI
| | - Catharine Song
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sompop Bencharit
- Department of Prosthodontics, School of Dentistry, University of North Carolina at Chapel Hill
| | - Ricardo Teles
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC.,General Oral Health Clinic, School of Dentistry, University of North Carolina at Chapel Hill
| | - Steven Offenbacher
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC.,General Oral Health Clinic, School of Dentistry, University of North Carolina at Chapel Hill
| |
Collapse
|
102
|
Shi G, Nehorai A. Robustness of meta-analyses in finding gene × environment interactions. PLoS One 2017; 12:e0171446. [PMID: 28362796 PMCID: PMC5375145 DOI: 10.1371/journal.pone.0171446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/20/2017] [Indexed: 12/31/2022] Open
Abstract
Meta-analyses that synthesize statistical evidence across studies have become important analytical tools for genetic studies. Inspired by the success of genome-wide association studies of the genetic main effect, researchers are searching for gene × environment interactions. Confounders are routinely included in the genome-wide gene × environment interaction analysis as covariates; however, this does not control for any confounding effects on the results if covariate × environment interactions are present. We carried out simulation studies to evaluate the robustness to the covariate × environment confounder for meta-regression and joint meta-analysis, which are two commonly used meta-analysis methods for testing the gene × environment interaction or the genetic main effect and interaction jointly. Here we show that meta-regression is robust to the covariate × environment confounder while joint meta-analysis is subject to the confounding effect with inflated type I error rates. Given vast sample sizes employed in genome-wide gene × environment interaction studies, non-significant covariate × environment interactions at the study level could substantially elevate the type I error rate at the consortium level. When covariate × environment confounders are present, type I errors can be controlled in joint meta-analysis by including the covariate × environment terms in the analysis at the study level. Alternatively, meta-regression can be applied, which is robust to potential covariate × environment confounders.
Collapse
Affiliation(s)
- Gang Shi
- State Key Laboratory of Integrated Services Networks, Xidian University, Xi’an, Shaanxi, China
- * E-mail:
| | - Arye Nehorai
- The Preston M. Green Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| |
Collapse
|
103
|
Castaldi PJ, Cho MH, Liang L, Silverman EK, Hersh CP, Rice K, Aschard H. Screening for interaction effects in gene expression data. PLoS One 2017; 12:e0173847. [PMID: 28301596 PMCID: PMC5354413 DOI: 10.1371/journal.pone.0173847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/27/2017] [Indexed: 11/27/2022] Open
Abstract
Expression quantitative trait (eQTL) studies are a powerful tool for identifying genetic variants that affect levels of messenger RNA. Since gene expression is controlled by a complex network of gene-regulating factors, one way to identify these factors is to search for interaction effects between genetic variants and mRNA levels of transcription factors (TFs) and their respective target genes. However, identification of interaction effects in gene expression data pose a variety of methodological challenges, and it has become clear that such analyses should be conducted and interpreted with caution. Investigating the validity and interpretability of several interaction tests when screening for eQTL SNPs whose effect on the target gene expression is modified by the expression level of a transcription factor, we characterized two important methodological issues. First, we stress the scale-dependency of interaction effects and highlight that commonly applied transformation of gene expression data can induce or remove interactions, making interpretation of results more challenging. We then demonstrate that, in the setting of moderate to strong interaction effects on the order of what may be reasonably expected for eQTL studies, standard interaction screening can be biased due to heteroscedasticity induced by true interactions. Using simulation and real data analysis, we outline a set of reasonable minimum conditions and sample size requirements for reliable detection of variant-by-environment and variant-by-TF interactions using the heteroscedasticity consistent covariance-based approach.
Collapse
Affiliation(s)
- Peter J. Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Hugues Aschard
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| |
Collapse
|
104
|
Waken RJ, de las Fuentes L, Rao DC. A Review of the Genetics of Hypertension with a Focus on Gene-Environment Interactions. Curr Hypertens Rep 2017; 19:23. [PMID: 28283927 PMCID: PMC5647656 DOI: 10.1007/s11906-017-0718-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Here, we discuss the interpretation and modeling of gene-environment interactions in hypertension-related phenotypes, with a focus on the necessary assumptions and possible challenges. RECENT FINDINGS Recently, small cohort studies have discovered several novel genetic variants associated with hypertension-related phenotypes through modeling gene-environment interactions. Several consortia-based meta-analytic efforts have uncovered many novel genetic variants in hypertension without modeling interaction terms, giving promise to future meta-analytic efforts that incorporate gene-environment interactions. Heritability studies and genome-wide association studies have established that hypertension, a prevalent cardiovascular disease, has a genetic component that may be modulated by the environment (such as lifestyle factors). This review includes a discussion of known genetic associations for hypertension/blood pressure, including those resulting from the incorporation of gene-environmental interaction modeling.
Collapse
Affiliation(s)
- R J Waken
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, 660 S. Euclid Ave, Campus Box 8067, St. Louis, MO, 63110, USA.
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, 660 S. Euclid Ave, Campus Box 8067, St. Louis, MO, 63110, USA
- Division of Cardiology, Department of Medicine, 660 S. Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| | - D C Rao
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, 660 S. Euclid Ave, Campus Box 8067, St. Louis, MO, 63110, USA
| |
Collapse
|
105
|
Pardo LM, Li WQ, Hwang SJ, Verkouteren JAC, Hofman A, Uitterlinden AG, Kraft P, Turman C, Han J, Cho E, Murabito JM, Levy D, Qureshi AA, Nijsten T. Genome-Wide Association Studies of Multiple Keratinocyte Cancers. PLoS One 2017; 12:e0169873. [PMID: 28081215 PMCID: PMC5231365 DOI: 10.1371/journal.pone.0169873] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/22/2016] [Indexed: 11/18/2022] Open
Abstract
There is strong evidence for a role of environmental risk factors involved in susceptibility to develop multiple keratinocyte cancers (mKCs), but whether genes are also involved in mKCs susceptibility has not been thoroughly investigated. We investigated whether single nucleotide polymorphisms (SNPs) are associated with susceptibility for mKCs. A genome-wide association study (GWAS) of 1,666 cases with mKCs and 1,950 cases with single KC (sKCs; controls) from Harvard cohorts (the Nurses' Health Study [NHS], NHS II, and the Health Professionals Follow-Up Study) and the Framingham Heart Study was carried-out using over 8 million SNPs (stage-1). We sought to replicate the most significant statistical associations (p-value≤ 5.5x10-6) in an independent cohort of 574 mKCs and 872 sKCs from the Rotterdam Study. In the discovery stage, 40 SNPs with suggestive associations (p-value ≤5.5x10-6) were identified, with eight independent SNPs tagging all 40 SNPs. The most significant SNP was located at chromosome 9 (rs7468390; p-value = 3.92x10-7). In stage-2, none of these SNPs replicated and only two of them were associated with mKCs in the same direction in the combined meta-analysis. We tested the associations for 19 previously reported basal cell carcinoma-related SNPs (candidate gene association analysis), and found that rs1805007 (MC1R locus) was significantly associated with risk of mKCs (p-value = 2.80x10-4). Although the suggestive SNPs with susceptibility for mKCs were not replicated, we found that previously identified BCC variants may also be associated with mKC, which the most significant association (rs1805007) located at the MC1R gene.
Collapse
Affiliation(s)
- Luba M. Pardo
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Wen-Qing Li
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence Rhode Island, United State of America
- Department of Epidemiology, School of Public Health, Brown University, Rhode Island, United State of America
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United State of America
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda Maryland MD United State of America
| | | | - Albert Hofman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United State of America
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA, United State of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United State of America
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, MA, United State of America
| | - Jiali Han
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United State of America
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, United State of America
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, United State of America
| | - Eunyoung Cho
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence Rhode Island, United State of America
- Department of Epidemiology, School of Public Health, Brown University, Rhode Island, United State of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United State of America
| | - Joanne M. Murabito
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United State of America
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United State of America
| | - Daniel Levy
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United State of America
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda Maryland MD United State of America
| | - Abrar A. Qureshi
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence Rhode Island, United State of America
- Department of Epidemiology, School of Public Health, Brown University, Rhode Island, United State of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United State of America
- * E-mail: (TN); (AAQ)
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- * E-mail: (TN); (AAQ)
| |
Collapse
|
106
|
Li X, Cornelis MC, Liang L, Song F, De Vivo I, Giovannucci E, Tang JY, Han J. A genome-wide analysis of gene-caffeine consumption interaction on basal cell carcinoma. Carcinogenesis 2016; 37:1138-1143. [PMID: 27797824 PMCID: PMC5137266 DOI: 10.1093/carcin/bgw107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/16/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022] Open
Abstract
Animal models have suggested that oral or topical administration of caffeine could inhibit ultraviolet-induced carcinogenesis via the ataxia telangiectasia and rad3 (ATR)-related apoptosis. Previous epidemiological studies have demonstrated that increased caffeine consumption is associated with reduced risk of basal cell carcinoma (BCC). To identify common genetic markers that may modify this association, we tested gene-caffeine intake interaction on BCC risk in a genome-wide analysis. We included 3383 BCC cases and 8528 controls of European ancestry from the Nurses' Health Study and Health Professionals Follow-up Study. Single nucleotide polymorphism (SNP) rs142310826 near the NEIL3 gene showed a genome-wide significant interaction with caffeine consumption (P = 1.78 × 10-8 for interaction) on BCC risk. There was no gender difference for this interaction (P = 0.64 for heterogeneity). NEIL3, a gene belonging to the base excision DNA repair pathway, encodes a DNA glycosylase that recognizes and removes lesions produced by oxidative stress. In addition, we identified several loci with P value for interaction <5 × 10-7 in gender-specific analyses (P for heterogeneity between genders < 0.001) including those mapping to the genes LRRTM4, ATF3 and DCLRE1C in women and POTEA in men. Finally, we tested the associations between caffeine consumption-related SNPs reported by previous genome-wide association studies and risk of BCC, both individually and jointly, but found no significant association. In sum, we identified a DNA repair gene that could be involved in caffeine-mediated skin tumor inhibition. Further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Xin Li
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA 02115, USA
| | - Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago IL 60611, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA 02115, USA
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Hospital and Institute, Tianjin 300060, China
- National Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jean Y Tang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA 94063, USA
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
- Center for Pharmacoepidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
107
|
Vaysse A, Fang S, Brossard M, Wei Q, Chen WV, Mohamdi H, Vincent-Fetita L, Margaritte-Jeannin P, Lavielle N, Maubec E, Lathrop M, Avril MF, Amos CI, Lee JE, Demenais F. A comprehensive genome-wide analysis of melanoma Breslow thickness identifies interaction between CDC42 and SCIN genetic variants. Int J Cancer 2016; 139:2012-20. [PMID: 27347659 PMCID: PMC5116391 DOI: 10.1002/ijc.30245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/07/2016] [Indexed: 12/23/2022]
Abstract
Breslow thickness (BT) is a major prognostic factor of cutaneous melanoma (CM), the most fatal skin cancer. The genetic component of BT has only been explored by candidate gene studies with inconsistent results. Our objective was to uncover the genetic factors underlying BT using an hypothesis-free genome-wide approach. Our analysis strategy integrated a genome-wide association study (GWAS) of single nucleotide polymorphisms (SNPs) for BT followed by pathway analysis of GWAS outcomes using the gene-set enrichment analysis (GSEA) method and epistasis analysis within BT-associated pathways. This strategy was applied to two large CM datasets with Hapmap3-imputed SNP data: the French MELARISK study for discovery (966 cases) and the MD Anderson Cancer Center study (1,546 cases) for replication. While no marginal effect of individual SNPs was revealed through GWAS, three pathways, defined by gene ontology (GO) categories were significantly enriched in genes associated with BT (false discovery rate ≤5% in both studies): hormone activity, cytokine activity and myeloid cell differentiation. Epistasis analysis, within each significant GO, identified a statistically significant interaction between CDC42 and SCIN SNPs (pmeta-int =2.2 × 10(-6) , which met the overall multiple-testing corrected threshold of 2.5 × 10(-6) ). These two SNPs (and proxies) are strongly associated with CDC42 and SCIN gene expression levels and map to regulatory elements in skin cells. This interaction has important biological relevance since CDC42 and SCIN proteins have opposite effects in actin cytoskeleton organization and dynamics, a key mechanism underlying melanoma cell migration and invasion.
Collapse
Affiliation(s)
- Amaury Vaysse
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Myriam Brossard
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center and Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Wei V. Chen
- Laboratory Informatics System, Department of Clinical Applications & Support, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Hamida Mohamdi
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Patricia Margaritte-Jeannin
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Nolwenn Lavielle
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Eve Maubec
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- AP-HP, Service de Dermatologie, Hôpital Avicenne et Université Paris 13, Bobigny, France
| | - Mark Lathrop
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | | | - Christopher I. Amos
- Department of Community and Family Medicine, Geisel College of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Florence Demenais
- INSERM, Genetic Variation and Human Diseases Unit, UMR-946, Paris, France
- Institut Universitaire d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
108
|
Brandariz SP, González Reymúndez A, Lado B, Malosetti M, Garcia AAF, Quincke M, von Zitzewitz J, Castro M, Matus I, del Pozo A, Castro AJ, Gutiérrez L. Ascertainment bias from imputation methods evaluation in wheat. BMC Genomics 2016; 17:773. [PMID: 27716058 PMCID: PMC5050639 DOI: 10.1186/s12864-016-3120-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/23/2016] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Whole-genome genotyping techniques like Genotyping-by-sequencing (GBS) are being used for genetic studies such as Genome-Wide Association (GWAS) and Genomewide Selection (GS), where different strategies for imputation have been developed. Nevertheless, imputation error may lead to poor performance (i.e. smaller power or higher false positive rate) when complete data is not required as it is for GWAS, and each marker is taken at a time. The aim of this study was to compare the performance of GWAS analysis for Quantitative Trait Loci (QTL) of major and minor effect using different imputation methods when no reference panel is available in a wheat GBS panel. RESULTS In this study, we compared the power and false positive rate of dissecting quantitative traits for imputed and not-imputed marker score matrices in: (1) a complete molecular marker barley panel array, and (2) a GBS wheat panel with missing data. We found that there is an ascertainment bias in imputation method comparisons. Simulating over a complete matrix and creating missing data at random proved that imputation methods have a poorer performance. Furthermore, we found that when QTL were simulated with imputed data, the imputation methods performed better than the not-imputed ones. On the other hand, when QTL were simulated with not-imputed data, the not-imputed method and one of the imputation methods performed better for dissecting quantitative traits. Moreover, larger differences between imputation methods were detected for QTL of major effect than QTL of minor effect. We also compared the different marker score matrices for GWAS analysis in a real wheat phenotype dataset, and we found minimal differences indicating that imputation did not improve the GWAS performance when a reference panel was not available. CONCLUSIONS Poorer performance was found in GWAS analysis when an imputed marker score matrix was used, no reference panel is available, in a wheat GBS panel.
Collapse
Affiliation(s)
- Sofía P. Brandariz
- Statistics Department, Facultad de Agronomía, Universidad de la República, Garzón 780, Montevideo, 12900 Uruguay
| | - Agustín González Reymúndez
- Statistics Department, Facultad de Agronomía, Universidad de la República, Garzón 780, Montevideo, 12900 Uruguay
| | - Bettina Lado
- Statistics Department, Facultad de Agronomía, Universidad de la República, Garzón 780, Montevideo, 12900 Uruguay
| | - Marcos Malosetti
- Biometris - Applied Statistics, Department of Plant Science, Wageningen University and Research Center, P.O. Box 16, 6700 AA Wageningen, Netherlands
| | - Antonio Augusto Franco Garcia
- Departamento de Ciências Exatas, Escola Superior de Agricultura “Luiz de Queiroz” (ESALQ), Universidade de São Paulo (USP), CP 9, CEP 13400-970 Piracicaba, SP Brazil
| | - Martín Quincke
- Programa Nacional de Investigación Cultivos de Secano, Instituto Nacional de investigación Agropecuaria, Est. Exp. La Estanzuela, Colonia, 70000 Uruguay
| | | | - Marina Castro
- Programa Nacional de Investigación Cultivos de Secano, Instituto Nacional de investigación Agropecuaria, Est. Exp. La Estanzuela, Colonia, 70000 Uruguay
| | - Iván Matus
- Instituto de Investigaciones Agropecuarias, Centro Regional de Investigación Quilamapu, Casilla 426, Chillán, Chile
| | - Alejandro del Pozo
- Facultad de Ciencias Agrarias, Universidad de Talca, Casilla 747, Talca, Chile
| | - Ariel J. Castro
- Department of Plant Production, Facultad de Agronomía, Universidad de la República, Ruta 3, Km.363, Paysandú, 60000 Uruguay
| | - Lucía Gutiérrez
- Statistics Department, Facultad de Agronomía, Universidad de la República, Garzón 780, Montevideo, 12900 Uruguay
- Department of Agronomy, University of Wisconsin-Madison, 1575 Linden Dr, Madison, WI 53706 USA
| |
Collapse
|
109
|
An Adaptive Fisher's Combination Method for Joint Analysis of Multiple Phenotypes in Association Studies. Sci Rep 2016; 6:34323. [PMID: 27694844 PMCID: PMC5046106 DOI: 10.1038/srep34323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022] Open
Abstract
Currently, the analyses of most genome-wide association studies (GWAS) have been performed on a single phenotype. There is increasing evidence showing that pleiotropy is a widespread phenomenon in complex diseases. Therefore, using only one single phenotype may lose statistical power to identify the underlying genetic mechanism. There is an increasing need to develop and apply powerful statistical tests to detect association between multiple phenotypes and a genetic variant. In this paper, we develop an Adaptive Fisher’s Combination (AFC) method for joint analysis of multiple phenotypes in association studies. The AFC method combines p-values obtained in standard univariate GWAS by using the optimal number of p-values which is determined by the data. We perform extensive simulations to evaluate the performance of the AFC method and compare the power of our method with the powers of TATES, Tippett’s method, Fisher’s combination test, MANOVA, MultiPhen, and SUMSCORE. Our simulation studies show that the proposed method has correct type I error rates and is either the most powerful test or comparable with the most powerful test. Finally, we illustrate our proposed methodology by analyzing whole-genome genotyping data from a lung function study.
Collapse
|
110
|
Amin N, Allebrandt KV, van der Spek A, Müller-Myhsok B, Hek K, Teder-Laving M, Hayward C, Esko T, van Mill JG, Mbarek H, Watson NF, Melville SA, Del Greco FM, Byrne EM, Oole E, Kolcic I, Chen TH, Evans DS, Coresh J, Vogelzangs N, Karjalainen J, Willemsen G, Gharib SA, Zgaga L, Mihailov E, Stone KL, Campbell H, Brouwer RWW, Demirkan A, Isaacs A, Dogas Z, Marciante KD, Campbell S, Borovecki F, Luik AI, Li M, Hottenga JJ, Huffman JE, van den Hout MCGN, Cummings SR, Aulchenko YS, Gehrman PR, Uitterlinden AG, Wichmann HE, Müller-Nurasyid M, Fehrmann RSN, Montgomery GW, Hofman A, Kao WHL, Oostra BA, Wright AF, Vink JM, Wilson JF, Pramstaller PP, Hicks AA, Polasek O, Punjabi NM, Redline S, Psaty BM, Heath AC, Merrow M, Tranah GJ, Gottlieb DJ, Boomsma DI, Martin NG, Rudan I, Tiemeier H, van IJcken WFJ, Penninx BW, Metspalu A, Meitinger T, Franke L, Roenneberg T, van Duijn CM. Genetic variants in RBFOX3 are associated with sleep latency. Eur J Hum Genet 2016; 24:1488-95. [PMID: 27142678 PMCID: PMC5027680 DOI: 10.1038/ejhg.2016.31] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 01/13/2016] [Accepted: 02/01/2016] [Indexed: 01/30/2023] Open
Abstract
Time to fall asleep (sleep latency) is a major determinant of sleep quality. Chronic, long sleep latency is a major characteristic of sleep-onset insomnia and/or delayed sleep phase syndrome. In this study we aimed to discover common polymorphisms that contribute to the genetics of sleep latency. We performed a meta-analysis of genome-wide association studies (GWAS) including 2 572 737 single nucleotide polymorphisms (SNPs) established in seven European cohorts including 4242 individuals. We found a cluster of three highly correlated variants (rs9900428, rs9907432 and rs7211029) in the RNA-binding protein fox-1 homolog 3 gene (RBFOX3) associated with sleep latency (P-values=5.77 × 10(-08), 6.59 × 10(-)(08) and 9.17 × 10(-)(08)). These SNPs were replicated in up to 12 independent populations including 30 377 individuals (P-values=1.5 × 10(-)(02), 7.0 × 10(-)(03) and 2.5 × 10(-)(03); combined meta-analysis P-values=5.5 × 10(-07), 5.4 × 10(-07) and 1.0 × 10(-07)). A functional prediction of RBFOX3 based on co-expression with other genes shows that this gene is predominantly expressed in brain (P-value=1.4 × 10(-316)) and the central nervous system (P-value=7.5 × 10(-)(321)). The predicted function of RBFOX3 based on co-expression analysis with other genes shows that this gene is significantly involved in the release cycle of neurotransmitters including gamma-aminobutyric acid and various monoamines (P-values<2.9 × 10(-11)) that are crucial in triggering the onset of sleep. To conclude, in this first large-scale GWAS of sleep latency we report a novel association of variants in RBFOX3 gene. Further, a functional prediction of RBFOX3 supports the involvement of RBFOX3 with sleep latency.
Collapse
Affiliation(s)
- Najaf Amin
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Karla V Allebrandt
- Institute of Medical Psychology, Ludwig-Maximilians-University, Munich, Germany
| | - Ashley van der Spek
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Karin Hek
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maris Teder-Laving
- Estonian Genome Center, University of Tartu and Estonian Biocenter, Tartu, Estonia
| | - Caroline Hayward
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu and Estonian Biocenter, Tartu, Estonia
| | - Josine G van Mill
- Department of Psychiatry, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Hamdi Mbarek
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Nathaniel F Watson
- Department of Neurology, University of Washington, Seattle, WA, USA
- University of Washington Medicine Sleep Center, Seattle, WA, USA
| | - Scott A Melville
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Fabiola M Del Greco
- Center for Biomedicine, European Academy of Bolzano, Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Enda M Byrne
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
- Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - Edwin Oole
- Center for Biomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ivana Kolcic
- School of Medicine, University of Split, Split, Croatia
| | - Ting-hsu Chen
- VA Boston Healthcare System, Boston University, Boston, MA, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Josef Coresh
- Departments of Epidemiology, Biostatistics, and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole Vogelzangs
- Department of Psychiatry, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Juha Karjalainen
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Sina A Gharib
- University of Washington Medicine Sleep Center, Seattle, WA, USA
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Lina Zgaga
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu and Estonian Biocenter, Tartu, Estonia
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Rutger WW Brouwer
- Center for Biomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ayse Demirkan
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Aaron Isaacs
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Zoran Dogas
- Department of Neuroscience and Sleep Medicine Centre, University of Split School of Medicine, Split, Croatia
| | - Kristin D Marciante
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan Campbell
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
| | - Fran Borovecki
- Centre for Functional Genomics and Department of Neurology, Faculty of Medicine, University of Zagreb, Zagreb, Croatia
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Man Li
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Jouke Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Jennifer E Huffman
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
| | | | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Yurii S Aulchenko
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Philip R Gehrman
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing and National Genomics Initiative, Leiden, The Netherlands
| | - Heinz-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum Munich-German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University and Klinikum Grosshadern, Munich, Germany
- Institute of Medical Statistics and Epidemiology, Technical University Munich, Munich, Germany
| | - Martina Müller-Nurasyid
- Institute of Epidemiology I, Helmholtz Zentrum Munich-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Rudolf SN Fehrmann
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | | | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wen Hong Linda Kao
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Ben A Oostra
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Alan F Wright
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
| | - Jacqueline M Vink
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - James F Wilson
- Medical Research Council, Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Peter P Pramstaller
- Center for Biomedicine, European Academy of Bolzano, Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Andrew A Hicks
- Center for Biomedicine, European Academy of Bolzano, Bolzano, Italy - Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Ozren Polasek
- School of Medicine, University of Split, Split, Croatia
- Centre for Global Health, University of Split School of Medicine, Split, Croatia
| | - Naresh M Punjabi
- Department of Pulmonary Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital and Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA, USA
| | - Andrew C Heath
- Department of Psychiatry, Washington University, St Louis, MO, USA
| | - Martha Merrow
- Institute of Medical Psychology, Ludwig-Maximilians-University, Munich, Germany
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Daniel J Gottlieb
- Department of Medicine, Brigham and Women's Hospital and Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | | | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus MC, Rotterdam, The Netherlands
| | | | - Brenda W Penninx
- Department of Psychiatry, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu and Estonian Biocenter, Tartu, Estonia
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Techinsche Universität München, München, Germany
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Till Roenneberg
- Institute of Medical Psychology, Ludwig-Maximilians-University, Munich, Germany
| | - Cornelia M van Duijn
- Unit of Genetic Epidemiology, Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing and National Genomics Initiative, Leiden, The Netherlands
- Centre for Medical Systems Biology, Leiden, The Netherlands
| |
Collapse
|
111
|
Tenaillon MI, Manicacci D, Nicolas SD, Tardieu F, Welcker C. Testing the link between genome size and growth rate in maize. PeerJ 2016; 4:e2408. [PMID: 27651994 PMCID: PMC5018661 DOI: 10.7717/peerj.2408] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/04/2016] [Indexed: 11/20/2022] Open
Abstract
Little is known about the factors driving within species Genome Size (GS) variation. GS may be shaped indirectly by natural selection on development and adaptative traits. Because GS variation is particularly pronounced in maize, we have sampled 83 maize inbred lines from three well described genetic groups adapted to contrasted climate conditions: inbreds of tropical origin, Flint inbreds grown in temperate climates, and Dent inbreds distributed in the Corn Belt. As a proxy for growth rate, we measured the Leaf Elongation Rate maximum during nighttime (LERmax) as well as GS in all inbred lines. In addition we combined available and new nucleotide polymorphism data at 29,090 sites to characterize the genetic structure of our panel. We found significant variation for both LERmax and GS among groups defined by our genetic structuring. Tropicals displayed larger GS than Flints while Dents exhibited intermediate values. LERmax followed the opposite trend with greater growth rate in Flints than in Tropicals. In other words, LERmax and GS exhibited a significantly negative correlation (r = − 0.27). However, this correlation was driven by among-group variation rather than within-group variation—it was no longer significant after controlling for structure and kinship among inbreds. Our results indicate that selection on GS may have accompanied ancient maize diffusion from its center of origin, with large DNA content excluded from temperate areas. Whether GS has been targeted by more intense selection during modern breeding within groups remains an open question.
Collapse
Affiliation(s)
- Maud I Tenaillon
- Génétique Quantitative et Evolution-Le Moulon, INRA-Université Paris-Sud-CNRS-AgroParisTech, Université Paris-Saclay , Gif-sur-Yvette , France
| | - Domenica Manicacci
- Génétique Quantitative et Evolution-Le Moulon, INRA-Université Paris-Sud-CNRS-AgroParisTech, Université Paris-Saclay , Gif-sur-Yvette , France
| | - Stéphane D Nicolas
- Génétique Quantitative et Evolution-Le Moulon, INRA-Université Paris-Sud-CNRS-AgroParisTech, Université Paris-Saclay , Gif-sur-Yvette , France
| | - Francois Tardieu
- Ecophysiologie des Plantes sous Stress Environnementaux, INRA , Montpellier , France
| | - Claude Welcker
- Ecophysiologie des Plantes sous Stress Environnementaux, INRA , Montpellier , France
| |
Collapse
|
112
|
Hill-Burns EM, Ross OA, Wissemann WT, Soto-Ortolaza AI, Zareparsi S, Siuda J, Lynch T, Wszolek ZK, Silburn PA, Mellick GD, Ritz B, Scherzer CR, Zabetian CP, Factor SA, Breheny PJ, Payami H. Identification of genetic modifiers of age-at-onset for familial Parkinson's disease. Hum Mol Genet 2016; 25:3849-3862. [PMID: 27402877 PMCID: PMC5216611 DOI: 10.1093/hmg/ddw206] [Citation(s) in RCA: 40] [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: 02/24/2016] [Revised: 06/15/2016] [Accepted: 06/23/2016] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease (PD) is the most common cause of neurodegenerative movement disorder and the second most common cause of dementia. Genes are thought to have a stronger effect on age-at-onset of PD than on risk, yet there has been a phenomenal success in identifying risk loci but not age-at-onset modifiers. We conducted a genome-wide study for age-at-onset. We analysed familial and non-familial PD separately, per prior evidence for strong genetic effect on age-at-onset in familial PD. GWAS was conducted in 431 unrelated PD individuals with at least one affected relative (familial PD) and 1544 non-familial PD from the NeuroGenetics Research Consortium (NGRC); an additional 737 familial PD and 2363 non-familial PD were used for replication. In familial PD, two signals were detected and replicated robustly: one mapped to LHFPL2 on 5q14.1 (PNGRC = 3E-8, PReplication = 2E-5, PNGRC + Replication = 1E-11), the second mapped to TPM1 on 15q22.2 (PNGRC = 8E-9, PReplication = 2E-4, PNGRC + Replication = 9E-11). The variants that were associated with accelerated onset had low frequencies (<0.02). The LHFPL2 variant was associated with earlier onset by 12.33 [95% CI: 6.2; 18.45] years in NGRC, 8.03 [2.95; 13.11] years in replication, and 9.79 [5.88; 13.70] years in the combined data. The TPM1 variant was associated with earlier onset by 15.30 [8.10; 22.49] years in NGRC, 9.29 [1.79; 16.79] years in replication, and 12.42 [7.23; 17.61] years in the combined data. Neither LHFPL2 nor TPM1 was associated with age-at-onset in non-familial PD. LHFPL2 (function unknown) is overexpressed in brain tumours. TPM1 encodes a highly conserved protein that regulates muscle contraction, and is a tumour-suppressor gene.
Collapse
Affiliation(s)
- Erin M Hill-Burns
- Department of Neurology, University of Alabama at Birmingham, AL, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic Jacksonville, FL, USA
| | | | | | - Sepideh Zareparsi
- Department of Molecular and Medical Genetics, Oregon Health & Sciences University, Portland, OR, USA
| | - Joanna Siuda
- Department of Neurology, Medical University of Silesia, Katowice, Poland
| | - Timothy Lynch
- Dublin Neurological Institute at the Mater Misericordiae University Hospital, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Ireland
| | | | - Peter A Silburn
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - George D Mellick
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health and Neurology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Clemens R Scherzer
- The Neurogenomics Laboratory, Harvard Medical School and Brigham & Women's Hospital, Cambridge, MA, USA
| | - Cyrus P Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA, USA
| | - Stewart A Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, AL, USA
- Center for Genomic Medicine, HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| |
Collapse
|
113
|
Wyszynski A, Hong CC, Lam K, Michailidou K, Lytle C, Yao S, Zhang Y, Bolla MK, Wang Q, Dennis J, Hopper JL, Southey MC, Schmidt MK, Broeks A, Muir K, Lophatananon A, Fasching PA, Beckmann MW, Peto J, Dos-Santos-Silva I, Sawyer EJ, Tomlinson I, Burwinkel B, Marme F, Guénel P, Truong T, Bojesen SE, Nordestgaard BG, González-Neira A, Benitez J, Neuhausen SL, Brenner H, Dieffenbach AK, Meindl A, Schmutzler RK, Brauch H, Nevanlinna H, Khan S, Matsuo K, Ito H, Dörk T, Bogdanova NV, Lindblom A, Margolin S, Mannermaa A, Kosma VM, Wu AH, Van Den Berg D, Lambrechts D, Wildiers H, Chang-Claude J, Rudolph A, Radice P, Peterlongo P, Couch FJ, Olson JE, Giles GG, Milne RL, Haiman CA, Henderson BE, Dumont M, Teo SH, Wong TY, Kristensen V, Zheng W, Long J, Winqvist R, Pylkäs K, Andrulis IL, Knight JA, Devilee P, Seynaeve C, García-Closas M, Figueroa J, Klevebring D, Czene K, Hooning MJ, van den Ouweland AMW, Darabi H, Shu XO, Gao YT, Cox A, Blot W, Signorello LB, Shah M, Kang D, Choi JY, Hartman M, Miao H, Hamann U, Jakubowska A, Lubinski J, Sangrajrang S, McKay J, Toland AE, Yannoukakos D, Shen CY, Wu PE, Swerdlow A, Orr N, et alWyszynski A, Hong CC, Lam K, Michailidou K, Lytle C, Yao S, Zhang Y, Bolla MK, Wang Q, Dennis J, Hopper JL, Southey MC, Schmidt MK, Broeks A, Muir K, Lophatananon A, Fasching PA, Beckmann MW, Peto J, Dos-Santos-Silva I, Sawyer EJ, Tomlinson I, Burwinkel B, Marme F, Guénel P, Truong T, Bojesen SE, Nordestgaard BG, González-Neira A, Benitez J, Neuhausen SL, Brenner H, Dieffenbach AK, Meindl A, Schmutzler RK, Brauch H, Nevanlinna H, Khan S, Matsuo K, Ito H, Dörk T, Bogdanova NV, Lindblom A, Margolin S, Mannermaa A, Kosma VM, Wu AH, Van Den Berg D, Lambrechts D, Wildiers H, Chang-Claude J, Rudolph A, Radice P, Peterlongo P, Couch FJ, Olson JE, Giles GG, Milne RL, Haiman CA, Henderson BE, Dumont M, Teo SH, Wong TY, Kristensen V, Zheng W, Long J, Winqvist R, Pylkäs K, Andrulis IL, Knight JA, Devilee P, Seynaeve C, García-Closas M, Figueroa J, Klevebring D, Czene K, Hooning MJ, van den Ouweland AMW, Darabi H, Shu XO, Gao YT, Cox A, Blot W, Signorello LB, Shah M, Kang D, Choi JY, Hartman M, Miao H, Hamann U, Jakubowska A, Lubinski J, Sangrajrang S, McKay J, Toland AE, Yannoukakos D, Shen CY, Wu PE, Swerdlow A, Orr N, Simard J, Pharoah PDP, Dunning AM, Chenevix-Trench G, Hall P, Bandera E, Amos C, Ambrosone C, Easton DF, Cole MD. An intergenic risk locus containing an enhancer deletion in 2q35 modulates breast cancer risk by deregulating IGFBP5 expression. Hum Mol Genet 2016; 25:3863-3876. [PMID: 27402876 PMCID: PMC5216618 DOI: 10.1093/hmg/ddw223] [Show More Authors] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/11/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the most diagnosed malignancy and the second leading cause of cancer mortality in females. Previous association studies have identified variants on 2q35 associated with the risk of breast cancer. To identify functional susceptibility loci for breast cancer, we interrogated the 2q35 gene desert for chromatin architecture and functional variation correlated with gene expression. We report a novel intergenic breast cancer risk locus containing an enhancer copy number variation (enCNV; deletion) located approximately 400Kb upstream to IGFBP5, which overlaps an intergenic ERα-bound enhancer that loops to the IGFBP5 promoter. The enCNV is correlated with modified ERα binding and monoallelic-repression of IGFBP5 following oestrogen treatment. We investigated the association of enCNV genotype with breast cancer in 1,182 cases and 1,362 controls, and replicate our findings in an independent set of 62,533 cases and 60,966 controls from 41 case control studies and 11 GWAS. We report a dose-dependent inverse association of 2q35 enCNV genotype (percopy OR = 0.68 95%CI 0.55-0.83, P = 0.0002; replication OR = 0.77 95% CI 0.73-0.82, P = 2.1 × 10-19) and identify 13 additional linked variants (r2 > 0.8) in the 20Kb linkage block containing the enCNV (P = 3.2 × 10-15 - 5.6 × 10-17). These associations were independent of previously reported 2q35 variants, rs13387042/rs4442975 and rs16857609, and were stronger for ER-positive than ER-negative disease. Together, these results suggest that 2q35 breast cancer risk loci may be mediating their effect through IGFBP5.
Collapse
Affiliation(s)
- Asaf Wyszynski
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | - Kyriaki Michailidou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Christian Lytle
- Molecular Biology Core Facility, Dartmouth College, Hanover, NH 03755 USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Yali Zhang
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Manjeet K Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Melissa C Southey
- Department of Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marjanka K Schmidt
- Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, 1066 CX Amsterdam, The Netherlands
| | - Annegien Broeks
- Wellcome Trust Centre for Human Genetics and Oxford Biomedical Research Centre, University of Oxford, OX3 7BN, UK
| | - Kenneth Muir
- Division of Health Sciences, Warwick Medical school, Warwick University, Coventry, CV4 7AL, UK
- Institute of Population Health, University of Manchester, Manchester, M13 9PL, UK
| | - Artitaya Lophatananon
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Peter A Fasching
- University Breast Center Franconia, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
- David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Matthias W Beckmann
- University Breast Center Franconia, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Elinor J Sawyer
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, SE1 9RT, UK
| | - Ian Tomlinson
- Wellcome Trust Centre for Human Genetics and Oxford Biomedical Research Centre, University of Oxford, OX3 7BN, UK
| | - Barbara Burwinkel
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
- Molecular Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frederik Marme
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
- National Center for Tumor Diseases, University of Heidelberg, 69120 Heidelberg, Germany
| | - Pascal Guénel
- Inserm (National Institute of Health and Medical Research), CESP (Center for Research in Epidemiology and Population Health), U1018, Environmental Epidemiology of Cancer, 94807 Villejuif, France
- University Paris-Sud, UMRS 1018, 94807 Villejuif, France
| | - Thérèse Truong
- Inserm (National Institute of Health and Medical Research), CESP (Center for Research in Epidemiology and Population Health), U1018, Environmental Epidemiology of Cancer, 94807 Villejuif, France
- University Paris-Sud, UMRS 1018, 94807 Villejuif, France
| | - Stig E Bojesen
- Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Børge G Nordestgaard
- Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Anna González-Neira
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Javier Benitez
- Human Genetics Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | | | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Aida Karina Dieffenbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Alfons Meindl
- Division of Gynaecology and Obstetrics, Technische Universität München, 81675 Munich, Germany
| | - Rita K Schmutzler
- Division of Molecular Gyneco-Oncology, Department of Gynaecology and Obstetrics, University Hospital of Cologne, 50931 Cologne, Germany
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, 50931 Cologne, Germany
- Center for Integrated Oncology (CIO), University Hospital of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 HUS, Finland
| | - Sofia Khan
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 HUS, Finland
| | - Keitaro Matsuo
- Department of Preventive Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, Japan
| | - Hidemi Ito
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Thilo Dörk
- Department of Obstetrics and Gynaecology, Hannover Medical School, 30625 Hannover, Germany
| | - Natalia V Bogdanova
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Sara Margolin
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Arto Mannermaa
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, FI-70211 Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Veli-Matti Kosma
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, FI-70211 Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - David Van Den Berg
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Diether Lambrechts
- Vesalius Research Center (VRC), VIB, 3000 Leuven, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| | - Hans Wildiers
- Multidisciplinary Breast Center, Department of General Medical Oncology, University Hospitals Leuven, B-3000 Leuven, Belgium
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori (INT), 20133 Milan, Italy
| | - Paolo Peterlongo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Graham G Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Roger L Milne
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Martine Dumont
- Centre Hospitalier Universitaire de Québec Research Center and Laval University, QC, G1V 4G2, Canada
| | - Soo Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, 47500 Subang Jaya, Selangor, Malaysia
- Breast Cancer Research Unit, University Malaya Cancer Research Institute, University Malaya Medical Centre (UMMC), 59100 Kuala Lumpur, Malaysia
| | - Tien Y Wong
- Singapore Eye Research Institute, National University of Singapore, Singapore 168751
| | - Vessela Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, N-0310 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo (UiO), 0450 Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), University of Oslo (UiO), 0450 Oslo, Norway
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Department of Clinical Chemistry and Biocenter Oulu, University of Oulu, NordLab Oulu/Oulu University Hospital, FI-90220 Oulu, Finland
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Department of Clinical Chemistry and Biocenter Oulu, University of Oulu, NordLab Oulu/Oulu University Hospital, FI-90220 Oulu, Finland
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Julia A Knight
- Prosserman Centre for Health Research, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Peter Devilee
- Department of Human Genetics & Department of Pathology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Caroline Seynaeve
- Family Cancer Clinic, Department of Medical Oncology, Erasmus MC-Daniel den Hoed Cancer Center, 3075 EA Rotterdam, The Netherlands
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, SM2 5NG, UK
- Breakthrough Breast Cancer Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jonine Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - Daniel Klevebring
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus University Medical Center, 3075 EA Rotterdam, The Netherlands
| | - Ans M W van den Ouweland
- Department of Clinical Genetics, Erasmus University Medical Center, 3075 EA Rotterdam, The Netherlands
| | - Hatef Darabi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Xuhui, Shanghai, China
| | - Angela Cox
- CRUK/YCR Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, Sheffield, S10 2RX, UK
| | - William Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- International Epidemiology Institute, Rockville, MD 20850, USA
| | - Lisa B Signorello
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Daehee Kang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 151-742, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Ji-Yeob Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 151-742, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Mikael Hartman
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117597
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore 117597
| | - Hui Miao
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117597
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, 70-115 Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, 70-115 Szczecin, Poland
| | | | - James McKay
- International Agency for Research on Cancer, 69372 Lyon, CEDEX 08, France
| | - Amanda E Toland
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, IRRP, National Centre for Scientific Research "Demokritos", Aghia Paraskevi Attikis, 153 10 Athens, Greece
| | - Chen-Yang Shen
- Taiwan Biobank, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- School of Public Health, China Medical University, Taichung 404, Taiwan
| | - Pei-Ei Wu
- Taiwan Biobank, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Anthony Swerdlow
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, SM2 5NG, UK
- Division of Breast Cancer Research, Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Nick Orr
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jacques Simard
- Centre Hospitalier Universitaire de Québec Research Center and Laval University, QC, G1V 4G2, Canada
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Georgia Chenevix-Trench
- Department of Genetics, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Elisa Bandera
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901 USA
| | - Chris Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Christine Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Michael D Cole
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
114
|
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: 69] [Impact Index Per Article: 7.7] [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
|
115
|
A thrifty variant in CREBRF strongly influences body mass index in Samoans. Nat Genet 2016; 48:1049-1054. [PMID: 27455349 PMCID: PMC5069069 DOI: 10.1038/ng.3620] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 06/15/2016] [Indexed: 12/14/2022]
Abstract
Samoans are a unique founder population with a high prevalence of obesity, making them well suited for identifying new genetic contributors to obesity. We conducted a genome-wide association study (GWAS) in 3,072 Samoans, discovered a variant, rs12513649, strongly associated with body mass index (BMI) (P = 5.3 × 10(-14)), and replicated the association in 2,102 additional Samoans (P = 1.2 × 10(-9)). Targeted sequencing identified a strongly associated missense variant, rs373863828 (p.Arg457Gln), in CREBRF (meta P = 1.4 × 10(-20)). Although this variant is extremely rare in other populations, it is common in Samoans (frequency of 0.259), with an effect size much larger than that of any other known common BMI risk variant (1.36-1.45 kg/m(2) per copy of the risk-associated allele). In comparison to wild-type CREBRF, the Arg457Gln variant when overexpressed selectively decreased energy use and increased fat storage in an adipocyte cell model. These data, in combination with evidence of positive selection of the allele encoding p.Arg457Gln, support a 'thrifty' variant hypothesis as a factor in human obesity.
Collapse
|
116
|
Common Genetic Variation in CYP17A1 and Response to Abiraterone Acetate in Patients with Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2016; 17:ijms17071097. [PMID: 27409606 PMCID: PMC4964473 DOI: 10.3390/ijms17071097] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 06/24/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023] Open
Abstract
Treatment with abiraterone acetate and prednisone (AA/P) prolongs survival in metastatic castration-resistant prostate cancer (mCRPC) patients. We evaluated the genetic variation in CYP17A1 as predictive of response to AA/P. A prospective collection of germline DNA prior to AA/P initiation and follow-up of a mCRPC cohort was performed. Five common single-nucleotide polymorphisms (SNPs) in CYP17A1 identified using a haplotype-based tagging algorithm were genotyped. Clinical outcomes included biochemical response and time to biochemical progression on AA/P. Logistic regression was used to assess the association between tag SNPs and biochemical response. Proportional hazards regression was used to assess the association between tag SNPs and time to biochemical progression. Odds or hazard ratio per minor allele were estimated and p-values below 0.05 were considered statistically significant. Germline DNA was successfully genotyped for four tag SNPs in 87 patients. The median age was 73 years (54-90); the median prostate-specific antigen was 66 ng/dL (0.1-99.9). A single SNP, rs2486758, was associated with lower odds of experiencing a biochemical response (Odds ratio 0.22, 95% confidence interval 0.07-0.63, p = 0.005) and a shorter time to biochemical progression (Hazard ratio 2.23, 95% confidence interval 1.39-3.56, p < 0.001). This tag SNP located in the promoter region of CYP17A1 will need further validation as a predictive biomarker for AA/P therapy.
Collapse
|
117
|
Mosteller M, Hosking L, Murphy K, Shen J, Song K, Nelson M, Ghosh S. No evidence of large genetic effects on steroid response in asthma patients. J Allergy Clin Immunol 2016; 139:797-803.e7. [PMID: 27523435 DOI: 10.1016/j.jaci.2016.05.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 05/17/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Inhaled corticosteroids (ICSs) are considered the most effective anti-inflammatory therapy for asthma control and management; however, there is substantial treatment response variability. OBJECTIVE We sought to identify genetic markers of ICS response by conducting the largest pharmacogenetic investigation to date in 2672 ICS-treated patients with asthma. METHODS Genotyping and imputation was performed in fluticasone furoate (FF) or fluticasone propionate-treated patients with asthma from 3 phase IIB and 4 phase IIIA randomized, double-blind, placebo-controlled, parallel group, multicenter studies. The primary end point analyzed was change in trough FEV1 (ΔFEV1) from baseline to 8 to 12 weeks of treatment. RESULTS More than 9.8 million common genetic variants (minor allele frequency ≥ 1%) were analyzed to test for association with ΔFEV1. No genetic variant met the prespecified threshold for statistical significance. CONCLUSIONS This study provides no evidence to confirm previously reported associations between candidate genetic variants and ICS response (ΔFEV1) in patients with asthma. In addition, no variant satisfied the criterion for genome-wide significance in our study. Common genetic variants are therefore unlikely to prove useful as predictive biomarkers of ICS response in patients with asthma.
Collapse
|
118
|
Fogh I, Lin K, Tiloca C, Rooney J, Gellera C, Diekstra FP, Ratti A, Shatunov A, van Es MA, Proitsi P, Jones A, Sproviero W, Chiò A, McLaughlin RL, Sorarù G, Corrado L, Stahl D, Del Bo R, Cereda C, Castellotti B, Glass JD, Newhouse S, Dobson R, Smith BN, Topp S, van Rheenen W, Meininger V, Melki J, Morrison KE, Shaw PJ, Leigh PN, Andersen PM, Comi GP, Ticozzi N, Mazzini L, D'Alfonso S, Traynor BJ, Van Damme P, Robberecht W, Brown RH, Landers JE, Hardiman O, Lewis CM, van den Berg LH, Shaw CE, Veldink JH, Silani V, Al-Chalabi A, Powell J. Association of a Locus in the CAMTA1 Gene With Survival in Patients With Sporadic Amyotrophic Lateral Sclerosis. JAMA Neurol 2016; 73:812-20. [PMID: 27244217 PMCID: PMC5556366 DOI: 10.1001/jamaneurol.2016.1114] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IMPORTANCE Amyotrophic lateral sclerosis (ALS) is a devastating adult-onset neurodegenerative disorder with a poor prognosis and a median survival of 3 years. However, a significant proportion of patients survive more than 10 years from symptom onset. OBJECTIVE To identify gene variants influencing survival in ALS. DESIGN, SETTING, AND PARTICIPANTS This genome-wide association study (GWAS) analyzed survival in data sets from several European countries and the United States that were collected by the Italian Consortium for the Genetics of ALS and the International Consortium on Amyotrophic Lateral Sclerosis Genetics. The study population included 4256 patients with ALS (3125 [73.4%] deceased) with genotype data extended to 7 174 392 variants by imputation analysis. Samples of DNA were collected from January 1, 1993, to December 31, 2009, and analyzed from March 1, 2014, to February 28, 2015. MAIN OUTCOMES AND MEASURES Cox proportional hazards regression under an additive model with adjustment for age at onset, sex, and the first 4 principal components of ancestry, followed by meta-analysis, were used to analyze data. Survival distributions for the most associated genetic variants were assessed by Kaplan-Meier analysis. RESULTS Among the 4256 patients included in the analysis (2589 male [60.8%] and 1667 female [39.2%]; mean [SD] age at onset, 59 [12] years), the following 2 novel loci were significantly associated with ALS survival: at 10q23 (rs139550538; P = 1.87 × 10-9) and in the CAMTA1 gene at 1p36 (rs2412208, P = 3.53 × 10-8). At locus 10q23, the adjusted hazard ratio for patients with the rs139550538 AA or AT genotype was 1.61 (95% CI, 1.38-1.89; P = 1.87 × 10-9), corresponding to an 8-month reduction in survival compared with TT carriers. For rs2412208 CAMTA1, the adjusted hazard ratio for patients with the GG or GT genotype was 1.17 (95% CI, 1.11-1.24; P = 3.53 × 10-8), corresponding to a 4-month reduction in survival compared with TT carriers. CONCLUSIONS AND RELEVANCE This GWAS robustly identified 2 loci at genome-wide levels of significance that influence survival in patients with ALS. Because ALS is a rare disease and prevention is not feasible, treatment that modifies survival is the most realistic strategy. Therefore, identification of modifier genes that might influence ALS survival could improve the understanding of the biology of the disease and suggest biological targets for pharmaceutical intervention. In addition, genetic risk scores for survival could be used as an adjunct to clinical trials to account for the genetic contribution to survival.
Collapse
Affiliation(s)
- Isabella Fogh
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Kuang Lin
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy
| | - James Rooney
- Academic Unit of Neurology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Frank P Diekstra
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Aleksey Shatunov
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Michael A van Es
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Petroula Proitsi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Ashley Jones
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - William Sproviero
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, ALS (Amyotrophic Lateral Sclerosis) Centre, University of Torino, Turin, Italy8Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Russell Lewis McLaughlin
- Population Genetics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Lucia Corrado
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy
| | - Daniel Stahl
- Department of Biostatistics, IoPPN, King's College London, London, England
| | - Roberto Del Bo
- Neurologic Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Cereda
- Laboratory of Experimental Neurobiology, IRCCS C. Mondino National Institute of Neurology Foundation, Pavia, Italy
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Steven Newhouse
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, IoPPN, King's College London, London, England17Department of Biostatistics, IoPPN, King's College London, London, England
| | - Richard Dobson
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, IoPPN, King's College London, London, England18NIHR Biomedical Research Unit in Dementia, King's College London, London, England
| | - Bradley N Smith
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Simon Topp
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Wouter van Rheenen
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincent Meininger
- Département des Maladies du Système Nerveux, Assistance Publique-Hôpitaux de Paris, Réseau SLA (Sclérose Latérale) Île de France, Hôpital Pitié-Salpêtrière, Paris, France
| | - Judith Melki
- Institut National de la Santé et de la Recherche Medicale Unité Mixte de Recherché-788 and University of Paris 11, Bicetre Hospital, Paris, France
| | - Karen E Morrison
- School of Clinical and Experimental Medicine, College of Medicine and Dentistry, University of Birmingham, Birmingham, England22Neurosciences Division, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham, England
| | - Pamela J Shaw
- Academic Neurology Unit, Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, England
| | - P Nigel Leigh
- Section of Neurology, Division of Medicine, Brighton and Sussex Medical School, Trafford Centre for Biomedical Research, University of Sussex, East Sussex, England
| | - Peter M Andersen
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany26Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Giacomo P Comi
- Neurologic Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Letizia Mazzini
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy27ALS Center Department of Neurology, Maggiore della Carità University Hospital, Novara, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, Flanders Instititue for Biotechnology, Vesalius Research Center, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium30Department of Neurology, University Hospitals Leuven, Leuven
| | - Wim Robberecht
- Department of Neurosciences, Experimental Neurology, Flanders Instititue for Biotechnology, Vesalius Research Center, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - Orla Hardiman
- Population Genetics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Cathryn M Lewis
- IoPPN Genomics and Biomarker Core, Translational Genetics Group, Medical Research Council Social, Genetic and Developmental Psychiatry Centre, King's College London, London, England33Department of Medical and Molecular Genetics, King's College London, Lon
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - John Powell
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| |
Collapse
|
119
|
Chen MM, O'Mara TA, Thompson DJ, Painter JN, Attia J, Black A, Brinton L, Chanock S, Chen C, Cheng TH, Cook LS, Crous-Bou M, Doherty J, Friedenreich CM, Garcia-Closas M, Gaudet MM, Gorman M, Haiman C, Hankinson SE, Hartge P, Henderson BE, Hodgson S, Holliday EG, Horn-Ross PL, Hunter DJ, Le Marchand L, Liang X, Lissowska J, Long J, Lu L, Magliocco AM, Martin L, McEvoy M, Olson SH, Orlow I, Pooler L, Prescott J, Rastogi R, Rebbeck TR, Risch H, Sacerdote C, Schumacher F, Wendy Setiawan V, Scott RJ, Sheng X, Shu XO, Turman C, Van Den Berg D, Wang Z, Weiss NS, Wentzensen N, Xia L, Xiang YB, Yang HP, Yu H, Zheng W, Pharoah PDP, Dunning AM, Tomlinson I, Easton DF, Kraft P, Spurdle AB, De Vivo I. GWAS meta-analysis of 16 852 women identifies new susceptibility locus for endometrial cancer. Hum Mol Genet 2016; 25:2612-2620. [PMID: 27008869 PMCID: PMC5868213 DOI: 10.1093/hmg/ddw092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/20/2022] Open
Abstract
Endometrial cancer is the most common gynecological malignancy in the developed world. Although there is evidence of genetic predisposition to the disease, most of the genetic risk remains unexplained. We present the meta-analysis results of four genome-wide association studies (4907 cases and 11 945 controls total) in women of European ancestry. We describe one new locus reaching genome-wide significance (P < 5 × 10 -8) at 6p22.3 (rs1740828; P = 2.29 × 10 -8, OR = 1.20), providing evidence of an additional region of interest for genetic susceptibility to endometrial cancer.
Collapse
Affiliation(s)
- Maxine M Chen
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Tracy A O'Mara
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Deborah J Thompson
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Jodie N Painter
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - John Attia
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, NSW 2305, Australia
- School of Medicine and Public Health, Centre for Clinical Epidemiology and Biostatistics, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Louise Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Chu Chen
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Timothy Ht Cheng
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Linda S Cook
- University of New Mexico, Albuquerque, NM 87131, USA
- Division of Cancer Care, Department of Population Health Research, Alberta Health Services, Calgary, AB, Canada
| | - Marta Crous-Bou
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer Doherty
- Geisel School of Medicine, Dartmouth College, Lebanon, NH 03755, USA
| | - Christine M Friedenreich
- Department of Cancer Epidemiology and Prevention Research, Cancer Control Alberta, Alberta Health Services, Calgary, AB, Canada
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Mia M Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30329, USA
| | - Maggie Gorman
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | | | - Susan E Hankinson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst MA 01003, USA
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Shirley Hodgson
- Department of Clinical Genetics, St George's, University of London, London SW17 0RE, UK
| | - Elizabeth G Holliday
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, NSW 2305, Australia
| | | | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | | | - Xiaolin Liang
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M Sklodowska-Curie Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Jirong Long
- Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt - Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN 27232, USA
| | - Lingeng Lu
- Yale University School of Public Health, New Haven, CT 06510, USA
| | | | - Lynn Martin
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Mark McEvoy
- School of Medicine and Public Health, Centre for Clinical Epidemiology and Biostatistics, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Sara H Olson
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irene Orlow
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Loreall Pooler
- University of Southern California, Los Angeles, CA 90033, USA
| | - Jennifer Prescott
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Radhai Rastogi
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Timothy R Rebbeck
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Harvey Risch
- Yale University School of Public Health, New Haven, CT 06510, USA
| | - Carlotta Sacerdote
- Center for Cancer Prevention (CPO-Piemonte), Turin, Italy
- Human Genetic Foundation (HuGeF), Turin, Italy
| | | | | | - Rodney J Scott
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, NSW 2305, Australia
- Hunter Area Pathology Service, John Hunter Hospital, Newcastle, NSW 2305, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Xin Sheng
- University of Southern California, Los Angeles, CA 90033, USA
| | - Xiao-Ou Shu
- Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt - Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN 27232, USA
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | | | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Noel S Weiss
- University of Washington, Seattle, WA 19024, USA
| | - Nicholas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Lucy Xia
- University of Southern California, Los Angeles, CA 90033, USA
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China and
| | - Hannah P Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Herbert Yu
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Wei Zheng
- Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt - Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN 27232, USA
| | - Paul D P Pharoah
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Alison M Dunning
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Ian Tomlinson
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Amanda B Spurdle
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Immaculata De Vivo
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA,
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
120
|
Sung YJ, Winkler TW, Manning AK, Aschard H, Gudnason V, Harris TB, Smith AV, Boerwinkle E, Brown MR, Morrison AC, Fornage M, Lin LA, Richard M, Bartz TM, Psaty BM, Hayward C, Polasek O, Marten J, Rudan I, Feitosa MF, Kraja AT, Province MA, Deng X, Fisher VA, Zhou Y, Bielak LF, Smith J, Huffman JE, Padmanabhan S, Smith BH, Ding J, Liu Y, Lohman K, Bouchard C, Rankinen T, Rice TK, Arnett D, Schwander K, Guo X, Palmas W, Rotter JI, Alfred T, Bottinger EP, Loos RJF, Amin N, Franco OH, van Duijn CM, Vojinovic D, Chasman DI, Ridker PM, Rose LM, Kardia S, Zhu X, Rice K, Borecki IB, Rao DC, Gauderman WJ, Cupples LA. An Empirical Comparison of Joint and Stratified Frameworks for Studying G × E Interactions: Systolic Blood Pressure and Smoking in the CHARGE Gene-Lifestyle Interactions Working Group. Genet Epidemiol 2016; 40:404-15. [PMID: 27230302 DOI: 10.1002/gepi.21978] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/08/2016] [Accepted: 04/04/2016] [Indexed: 01/11/2023]
Abstract
Studying gene-environment (G × E) interactions is important, as they extend our knowledge of the genetic architecture of complex traits and may help to identify novel variants not detected via analysis of main effects alone. The main statistical framework for studying G × E interactions uses a single regression model that includes both the genetic main and G × E interaction effects (the "joint" framework). The alternative "stratified" framework combines results from genetic main-effect analyses carried out separately within the exposed and unexposed groups. Although there have been several investigations using theory and simulation, an empirical comparison of the two frameworks is lacking. Here, we compare the two frameworks using results from genome-wide association studies of systolic blood pressure for 3.2 million low frequency and 6.5 million common variants across 20 cohorts of European ancestry, comprising 79,731 individuals. Our cohorts have sample sizes ranging from 456 to 22,983 and include both family-based and population-based samples. In cohort-specific analyses, the two frameworks provided similar inference for population-based cohorts. The agreement was reduced for family-based cohorts. In meta-analyses, agreement between the two frameworks was less than that observed in cohort-specific analyses, despite the increased sample size. In meta-analyses, agreement depended on (1) the minor allele frequency, (2) inclusion of family-based cohorts in meta-analysis, and (3) filtering scheme. The stratified framework appears to approximate the joint framework well only for common variants in population-based cohorts. We conclude that the joint framework is the preferred approach and should be used to control false positives when dealing with low-frequency variants and/or family-based cohorts.
Collapse
Affiliation(s)
- Yun Ju Sung
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Thomas W Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Alisa K Manning
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America.,Center for Human Genetics Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America.,Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Li-An Lin
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Traci M Bartz
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia.,Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jonathan Marten
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Igor Rudan
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Virginia A Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Jennifer Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Jennifer E Huffman
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Blair H Smith
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Kurt Lohman
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Los Angeles, United States of America
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Los Angeles, United States of America
| | - Treva K Rice
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Donna Arnett
- Department of Epidemiology, University of Alabama-Birmingham, Birmingham, Alabama, United States of America
| | - Karen Schwander
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - Xiuqing Guo
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Jerome I Rotter
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Tamuno Alfred
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America.,The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Oscar H Franco
- Cardiovascular Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Dina Vojinovic
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Daniel I Chasman
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Paul M Ridker
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Lynda M Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Sharon Kardia
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kenneth Rice
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America.,Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University, St. Louis, Missouri, United States of America
| | - W James Gauderman
- Department of Preventive Medicine, University of Southern California, Los Angeles, California, United States of America
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America.,Framingham Heart Study, Framingham, Massachusetts, United States of America
| |
Collapse
|
121
|
Abstract
Development of free/libre open source software is usually done by a community of people with an interest in the tool. For scientific software, however, this is less often the case. Most scientific software is written by only a few authors, often a student working on a thesis. Once the paper describing the tool has been published, the tool is no longer developed further and is left to its own device. Here we describe the broad, multidisciplinary community we formed around a set of tools for statistical genomics. The GenABEL project for statistical omics actively promotes open interdisciplinary development of statistical methodology and its implementation in efficient and user-friendly software under an open source licence. The software tools developed withing the project collectively make up the GenABEL suite, which currently consists of eleven tools. The open framework of the project actively encourages involvement of the community in all stages, from formulation of methodological ideas to application of software to specific data sets. A web forum is used to channel user questions and discussions, further promoting the use of the GenABEL suite. Developer discussions take place on a dedicated mailing list, and development is further supported by robust development practices including use of public version control, code review and continuous integration. Use of this open science model attracts contributions from users and developers outside the “core team”, facilitating agile statistical omics methodology development and fast dissemination.
Collapse
Affiliation(s)
- Lennart C Karssen
- PolyOmica, Groningen, 9722 HC, Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3000 CA, Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3000 CA, Netherlands
| | - Yurii S Aulchenko
- PolyOmica, Groningen, 9722 HC, Netherlands; Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, 630090, Russian Federation; Novosibirsk State University, Novosibirsk, 630090, Russian Federation; Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, UK
| |
Collapse
|
122
|
Offenbacher S, Divaris K, Barros SP, Moss KL, Marchesan JT, Morelli T, Zhang S, Kim S, Sun L, Beck JD, Laudes M, Munz M, Schaefer AS, North KE. Genome-wide association study of biologically informed periodontal complex traits offers novel insights into the genetic basis of periodontal disease. Hum Mol Genet 2016; 25:2113-2129. [PMID: 26962152 PMCID: PMC5062586 DOI: 10.1093/hmg/ddw069] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 02/19/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWAS) of chronic periodontitis (CP) defined by clinical criteria alone have had modest success to-date. Here, we refine the CP phenotype by supplementing clinical data with biological intermediates of microbial burden (levels of eight periodontal pathogens) and local inflammatory response (gingival crevicular fluid IL-1β) and derive periodontal complex traits (PCTs) via principal component analysis. PCTs were carried forward to GWAS (∼2.5 million markers) to identify PCT-associated loci among 975 European American adult participants of the Dental ARIC study. We sought to validate these findings for CP in the larger ARIC cohort (n = 821 participants with severe CP, 2031-moderate CP, 1914-healthy/mild disease) and an independent German sample including 717 aggressive periodontitis cases and 4210 controls. We identified six PCTs with distinct microbial community/IL-1β structures, although with overlapping clinical presentations. PCT1 was characterized by a uniformly high pathogen load, whereas PCT3 and PCT5 were dominated by Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis, respectively. We detected genome-wide significant signals for PCT1 (CLEC19A, TRA, GGTA2P, TM9SF2, IFI16, RBMS3), PCT4 (HPVC1) and PCT5 (SLC15A4, PKP2, SNRPN). Overall, the highlighted loci included genes associated with immune response and epithelial barrier function. With the exception of associations of BEGAIN with severe and UBE3D with moderate CP, no other loci were associated with CP in ARIC or aggressive periodontitis in the German sample. Although not associated with current clinically determined periodontal disease taxonomies, upon replication and mechanistic validation these candidate loci may highlight dysbiotic microbial community structures and altered inflammatory/immune responses underlying biological sub-types of CP.
Collapse
Affiliation(s)
- Steven Offenbacher
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Kimon Divaris
- Department of Pediatric Dentistry, UNC School of Dentistry, Chapel Hill, NC, USA Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Silvana P Barros
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Kevin L Moss
- Department of Dental Ecology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Julie T Marchesan
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Thiago Morelli
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Shaoping Zhang
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Steven Kim
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Lu Sun
- Department of Periodontology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - James D Beck
- Department of Dental Ecology, UNC School of Dentistry, Chapel Hill, NC, USA
| | - Matthias Laudes
- Clinic of Internal Medicine I, University Clinic Schleswig-Holstein, Kiel, Germany
| | - Matthias Munz
- Department of Periodontology, Institute of Dental, Oral and Maxillary Medicine, Charité-University Medicine Berlin, Berlin, Germany Institute of Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
| | - Arne S Schaefer
- Department of Periodontology, Institute of Dental, Oral and Maxillary Medicine, Charité-University Medicine Berlin, Berlin, Germany
| | - Kari E North
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| |
Collapse
|
123
|
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: 0.9] [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
|
124
|
Tracking Resilience to Infections by Mapping Disease Space. PLoS Biol 2016; 14:e1002436. [PMID: 27088359 PMCID: PMC4835107 DOI: 10.1371/journal.pbio.1002436] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/15/2016] [Indexed: 11/19/2022] Open
Abstract
Infected hosts differ in their responses to pathogens; some hosts are resilient and recover their original health, whereas others follow a divergent path and die. To quantitate these differences, we propose mapping the routes infected individuals take through “disease space.” We find that when plotting physiological parameters against each other, many pairs have hysteretic relationships that identify the current location of the host and predict the future route of the infection. These maps can readily be constructed from experimental longitudinal data, and we provide two methods to generate the maps from the cross-sectional data that is commonly gathered in field trials. We hypothesize that resilient hosts tend to take small loops through disease space, whereas nonresilient individuals take large loops. We support this hypothesis with experimental data in mice infected with Plasmodium chabaudi, finding that dying mice trace a large arc in red blood cells (RBCs) by reticulocyte space as compared to surviving mice. We find that human malaria patients who are heterozygous for sickle cell hemoglobin occupy a small area of RBCs by reticulocyte space, suggesting this approach can be used to distinguish resilience in human populations. This technique should be broadly useful in describing the in-host dynamics of infections in both model hosts and patients at both population and individual levels. This study shows that infections cause sick hosts to loop through disease space on their return back to health; resilient individuals take tiny loops through this space, minimizing the impact of the infection. When we get sick, we long for recovery; thus, a major goal of medicine is to promote resilience—the ability of a host to return to its original health following an infection. While in the laboratory we can study the response to infection with precise knowledge of inoculation time and dose, sick patients in the clinic do not have this information. This creates a problem because we can’t easily differentiate between patients who are early in the stages of infection that will develop severe disease from more disease-tolerant patients who present later in the infection. The distinction between these two types of patients is important, as the less disease-tolerant patient would require a more aggressive treatment regime. To determine where patients lie along the infection timeline, we charted “disease maps” that trace a patient’s route through “disease space.” We select symptoms that produce looping graphs as patients grow sick and recover. Using a mouse–malaria model, we demonstrate that less resilient individuals take wider loops through this space, representing a longer infection time with more severe symptoms. We find this looping behavior also applies to humans and suggest that people carrying the sickle cell trait are more resilient to malaria infections.
Collapse
|
125
|
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: 4.0] [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
|
126
|
Dunn EC, Wiste A, Radmanesh F, Almli LM, Gogarten SM, Sofer T, Faul JD, Kardia SL, Smith JA, Weir DR, Zhao W, Soare TW, Mirza SS, Hek K, Tiemeier HW, Goveas JS, Sarto GE, Snively BM, Cornelis M, Koenen KC, Kraft P, Purcell S, Ressler KJ, Rosand J, Wassertheil-Smoller S, Smoller JW. GENOME-WIDE ASSOCIATION STUDY (GWAS) AND GENOME-WIDE BY ENVIRONMENT INTERACTION STUDY (GWEIS) OF DEPRESSIVE SYMPTOMS IN AFRICAN AMERICAN AND HISPANIC/LATINA WOMEN. Depress Anxiety 2016; 33:265-80. [PMID: 27038408 PMCID: PMC4826276 DOI: 10.1002/da.22484] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have made little progress in identifying variants linked to depression. We hypothesized that examining depressive symptoms and considering gene-environment interaction (GxE) might improve efficiency for gene discovery. We therefore conducted a GWAS and genome-wide by environment interaction study (GWEIS) of depressive symptoms. METHODS Using data from the SHARe cohort of the Women's Health Initiative, comprising African Americans (n = 7,179) and Hispanics/Latinas (n = 3,138), we examined genetic main effects and GxE with stressful life events and social support. We also conducted a heritability analysis using genome-wide complex trait analysis (GCTA). Replication was attempted in four independent cohorts. RESULTS No SNPs achieved genome-wide significance for main effects in either discovery sample. The top signals in African Americans were rs73531535 (located 20 kb from GPR139, P = 5.75 × 10(-8) ) and rs75407252 (intronic to CACNA2D3, P = 6.99 × 10(-7) ). In Hispanics/Latinas, the top signals were rs2532087 (located 27 kb from CD38, P = 2.44 × 10(-7) ) and rs4542757 (intronic to DCC, P = 7.31 × 10(-7) ). In the GEWIS with stressful life events, one interaction signal was genome-wide significant in African Americans (rs4652467; P = 4.10 × 10(-10) ; located 14 kb from CEP350). This interaction was not observed in a smaller replication cohort. Although heritability estimates for depressive symptoms and stressful life events were each less than 10%, they were strongly genetically correlated (rG = 0.95), suggesting that common variation underlying self-reported depressive symptoms and stressful life event exposure, though modest on their own, were highly overlapping in this sample. CONCLUSIONS Our results underscore the need for larger samples, more GEWIS, and greater investigation into genetic and environmental determinants of depressive symptoms in minorities.
Collapse
Affiliation(s)
- Erin C. Dunn
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| | - Anna Wiste
- Center for Experimental Drugs and Diagnostics, Department of Psychiatry, Massachusetts General Hospital
| | - Farid Radmanesh
- Center for Human Genetic Research, Massachusetts General Hospital
- Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT
| | - Lynn M. Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | | | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Jessica D. Faul
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | | | - Jennifer A. Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - David R. Weir
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Thomas W. Soare
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| | - Saira S. Mirza
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Karin Hek
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Henning W. Tiemeier
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joseph S. Goveas
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gloria E. Sarto
- Center for Women's Health and Health Disparities Research, Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Beverly M. Snively
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Marilyn Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Karestan C. Koenen
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health
| | - Shaun Purcell
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kerry J. Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Jonathan Rosand
- Center for Human Genetic Research, Massachusetts General Hospital
- Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York
| | - Jordan W. Smoller
- Center for Human Genetic Research, Massachusetts General Hospital
- Department of Psychiatry, Harvard Medical School
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT
| |
Collapse
|
127
|
Adams HH, Ikram MA, Vernooij MW, van Dijk AC, Hofman A, Uitterlinden AG, van Duijn CM, Koudstaal PJ, Franco OH, van der Lugt A, Bos D. Heritability and Genome-Wide Association Analyses of Intracranial Carotid Artery Calcification. Stroke 2016; 47:912-7. [DOI: 10.1161/strokeaha.115.012248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/10/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Hieab H.H. Adams
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - M. Arfan Ikram
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Meike W. Vernooij
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Anouk C. van Dijk
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Albert Hofman
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - André G. Uitterlinden
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Cornelia M. van Duijn
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Peter J. Koudstaal
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Oscar H. Franco
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Aad van der Lugt
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| | - Daniel Bos
- From the Departments of Epidemiology (H.H.H.A., M.A.I., M.W.V., A.H., A.G.U., C.M.v.D., O.H.F., D.B.), Radiology (M.A.I., M.W.V., A.C.v.D., A.v.d.L., D.B.), Neurology (M.A.I., A.C.v.D., P.J.K.), and Internal Medicine (A.G.U.), Erasmus MC, Rotterdam, the Netherlands; and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (A.H.)
| |
Collapse
|
128
|
Genome-wide significant results identified for plasma apolipoprotein H levels in middle-aged and older adults. Sci Rep 2016; 6:23675. [PMID: 27030319 PMCID: PMC4814826 DOI: 10.1038/srep23675] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/07/2016] [Indexed: 11/09/2022] Open
Abstract
Apolipoprotein H (ApoH) is a multi-functional plasma glycoprotein that has been associated with negative health outcomes. ApoH levels have high heritability. We undertook a genome-wide association study of ApoH levels using the largest sample to date and replicated the results in an independent cohort (total N = 1,255). In the discovery phase, a meta-analysis of two cohorts, the Sydney Memory and Ageing Study (Sydney MAS) and the Older Australian Twins Study (OATS) (n = 942) revealed genome-wide significant results in or near the APOH gene on chromosome 17 (top SNP, rs7211380, p = 1 × 10−11). The results were replicated in an independent cohort, the Hunter Community Study (p < 0.002) (n = 313). Conditional and joint analysis (COJO) confirmed the association of the chromosomal 17 region with ApoH levels. The set of independent SNPs identified by COJO explained 23% of the variance. The relationships between the top SNPs and cardiovascular/lipid/cognition measures and diabetes were assessed in Sydney MAS, with suggestive results observed for diabetes and cognitive performance. However, replication of these results in the smaller OATS cohort was not found. This work provides impetus for future research to better understand the contribution of genetics to ApoH levels and its possible impacts on health.
Collapse
|
129
|
Thompson DJ, O'Mara TA, Glubb DM, Painter JN, Cheng T, Folkerd E, Doody D, Dennis J, Webb PM, Gorman M, Martin L, Hodgson S, Michailidou K, Tyrer JP, Maranian MJ, Hall P, Czene K, Darabi H, Li J, Fasching PA, Hein A, Beckmann MW, Ekici AB, Dörk T, Hillemanns P, Dürst M, Runnebaum I, Zhao H, Depreeuw J, Schrauwen S, Amant F, Goode EL, Fridley BL, Dowdy SC, Winham SJ, Salvesen HB, Trovik J, Njolstad TS, Werner HMJ, Ashton K, Proietto T, Otton G, Carvajal-Carmona L, Tham E, Liu T, Mints M, Scott RJ, McEvoy M, Attia J, Holliday EG, Montgomery GW, Martin NG, Nyholt DR, Henders AK, Hopper JL, Traficante N, Ruebner M, Swerdlow AJ, Burwinkel B, Brenner H, Meindl A, Brauch H, Lindblom A, Lambrechts D, Chang-Claude J, Couch FJ, Giles GG, Kristensen VN, Cox A, Bolla MK, Wang Q, Bojesen SE, Shah M, Luben R, Khaw KT, Pharoah PDP, Dunning AM, Tomlinson I, Dowsett M, Easton DF, Spurdle AB. CYP19A1 fine-mapping and Mendelian randomization: estradiol is causal for endometrial cancer. Endocr Relat Cancer 2016; 23:77-91. [PMID: 26574572 PMCID: PMC4697192 DOI: 10.1530/erc-15-0386] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 10/22/2015] [Accepted: 11/16/2015] [Indexed: 12/19/2022]
Abstract
Candidate gene studies have reported CYP19A1 variants to be associated with endometrial cancer and with estradiol (E2) concentrations. We analyzed 2937 single nucleotide polymorphisms (SNPs) in 6608 endometrial cancer cases and 37 925 controls and report the first genome wide-significant association between endometrial cancer and a CYP19A1 SNP (rs727479 in intron 2, P=4.8×10(-11)). SNP rs727479 was also among those most strongly associated with circulating E2 concentrations in 2767 post-menopausal controls (P=7.4×10(-8)). The observed endometrial cancer odds ratio per rs727479 A-allele (1.15, CI=1.11-1.21) is compatible with that predicted by the observed effect on E2 concentrations (1.09, CI=1.03-1.21), consistent with the hypothesis that endometrial cancer risk is driven by E2. From 28 candidate-causal SNPs, 12 co-located with three putative gene-regulatory elements and their risk alleles associated with higher CYP19A1 expression in bioinformatical analyses. For both phenotypes, the associations with rs727479 were stronger among women with a higher BMI (Pinteraction=0.034 and 0.066 respectively), suggesting a biologically plausible gene-environment interaction.
Collapse
Affiliation(s)
- Deborah J Thompson
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
| | - Tracy A O'Mara
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Dylan M Glubb
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Jodie N Painter
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Timothy Cheng
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Elizabeth Folkerd
- Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Deborah Doody
- Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Joe Dennis
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
| | - Penelope M Webb
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | | | - Maggie Gorman
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Lynn Martin
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Shirley Hodgson
- Department of Clinical Genetics, St George's Hospital Medical School, London, SW17 0RE, UK
| | | | - Kyriaki Michailidou
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
| | - Jonathan P Tyrer
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Mel J Maranian
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Hatef Darabi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Jingmei Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Peter A Fasching
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, 90095, USA
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Alexander Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Arif B Ekici
- Institute of Human Genetics, , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, 30625, Germany
| | - Peter Hillemanns
- Clinics of Gynaecology and Obstetrics, Hannover Medical School, Hannover, 30625, Germany
| | - Matthias Dürst
- Department of Gynaecology, Jena University Hospital – Friedrich Schiller University, Jena, 07743, Germany
| | - Ingo Runnebaum
- Department of Gynaecology, Jena University Hospital – Friedrich Schiller University, Jena, 07743, Germany
| | - Hui Zhao
- Vesalius Research Center, Leuven, 3000, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Jeroen Depreeuw
- Vesalius Research Center, Leuven, 3000, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University Hospitals Leuven, Leuven, 3000, Belgium
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospitals, KU Leuven – University of Leuven, Leuven, 3000, Belgium
| | - Stefanie Schrauwen
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospitals, KU Leuven – University of Leuven, Leuven, 3000, Belgium
| | - Frederic Amant
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospitals, KU Leuven – University of Leuven, Leuven, 3000, Belgium
| | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Brooke L Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, 66160, USA
| | - Sean C Dowdy
- Department of Obstetrics and Gynecology Division of Gynecologic Oncology Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Stacey J Winham
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Helga B Salvesen
- Department of Clinical Science, Centre for Cancerbiomarkers, The University of Bergen, Bergen, 5020, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, 5021, Norway
| | - Jone Trovik
- Department of Clinical Science, Centre for Cancerbiomarkers, The University of Bergen, Bergen, 5020, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, 5021, Norway
| | - Tormund S Njolstad
- Department of Clinical Science, Centre for Cancerbiomarkers, The University of Bergen, Bergen, 5020, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, 5021, Norway
| | - Henrica M J Werner
- Department of Clinical Science, Centre for Cancerbiomarkers, The University of Bergen, Bergen, 5020, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, 5021, Norway
| | - Katie Ashton
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, New South Wales, 2305, Australia
- Centre for Information Based Medicine, University of Newcastle, Newcastle, New South Wales, 2308, Australia
- School of Biomedical Sciences and Pharmacy, , University of Newcastle Newcastle, Newcastle, New South Wales, 2308, Australia
| | - Tony Proietto
- School of Medicine and Public Health, , University of Newcastle, Newcastle, Newcastle, New South Wales, 2308, Australia
| | - Geoffrey Otton
- School of Medicine and Public Health, , University of Newcastle, Newcastle, Newcastle, New South Wales, 2308, Australia
| | - Luis Carvajal-Carmona
- Grupo de investigación Citogenética, Filogenia y Evolución de Poblaciones, Universidad del Tolima, Ibagué, Tolima, Colombia
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, California, 95616, USA
| | - Emma Tham
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Tao Liu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Miriam Mints
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SE-171 77, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, SE-171 77, Sweden
| | - for RENDOCAS
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Rodney J Scott
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, New South Wales, 2305, Australia
- Centre for Information Based Medicine, University of Newcastle, Newcastle, New South Wales, 2308, Australia
- School of Biomedical Sciences and Pharmacy, , University of Newcastle Newcastle, Newcastle, New South Wales, 2308, Australia
- Hunter Area Pathology Service, John Hunter Hospital, Newcastle, New South Wales, 2305, Australia
| | - Mark McEvoy
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, 2305, Australia
| | - John Attia
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, New South Wales, 2305, Australia
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, 2305, Australia
| | - Elizabeth G Holliday
- Hunter Medical Research Institute, John Hunter Hospital, Newcastle, New South Wales, 2305, Australia
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, 2305, Australia
| | - Grant W Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Dale R Nyholt
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4006, Australia
| | - Anjali K Henders
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne victoria, Melbourne, Victoria, 3010, Australia
| | - Nadia Traficante
- PePeter MacCallum Cancer Center, The University of Melbourne, Melbourne, 3002, Australia
| | - for the AOCS Group
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91054, Germany
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, SM2 5NG, UK
- Division of Breast Cancer Research, Institute of Cancer Research, London, SM2 5NG, UK
| | - Barbara Burwinkel
- Department of Gynecology and Obstetrics, Molecular Biology of Breast Cancer, University of Heidelberg, Heidelberg, 69117, Germany
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, 69120, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Alfons Meindl
- Department of Obstetrics and Gynecology, Division of Tumor Genetics, Technical University of Munich, Munich, 80333, Germany
| | - Hiltrud Brauch
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- University of Tübingen, Tübingen, 72074, Germany
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Diether Lambrechts
- Vesalius Research Center, Leuven, 3000, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, 69120, Germany
| | - Fergus J Couch
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, 55905, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne victoria, Melbourne, Victoria, 3010, Australia
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, 3004, Australia
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, 0310, Norway
- Faculty of Medicine, The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, , University of Oslo, Oslo, 0316, Norway
- Department of Clinical Molecular Oncology, Division of Medicine, Akershus University Hospital, Lørenskog, 1478, Norway
| | - Angela Cox
- Department of Oncology, Sheffield Cancer Research, University of Sheffield, Sheffield, S10 2TN, UK
| | - Manjeet K Bolla
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
| | - Qin Wang
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
| | - Stig E Bojesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 1165, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, 2730, Denmark
| | - Mitul Shah
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Robert Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Kay-Tee Khaw
- MRC Centre for Nutritional Epidemiology in Cancer Prevention and Survival (CNC), University of Cambridge, Cambridge, CB1 8RN, UK
| | - Paul D P Pharoah
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Alison M Dunning
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Ian Tomlinson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Mitch Dowsett
- Academic Department of Biochemistry, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Amanda B Spurdle
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| |
Collapse
|
130
|
Bailey JNC, Loomis SJ, Kang JH, Allingham RR, Gharahkhani P, Khor CC, Burdon KP, Aschard H, Chasman DI, Igo RP, Hysi PG, Glastonbury CA, Ashley-Koch A, Brilliant M, Brown AA, Budenz DL, Buil A, Cheng CY, Choi H, Christen WG, Curhan G, De Vivo I, Fingert JH, Foster PJ, Fuchs C, Gaasterland D, Gaasterland T, Hewitt AW, Hu F, Hunter DJ, Khawaja AP, Lee RK, Li Z, Lichter PR, Mackey DA, McGuffin P, Mitchell P, Moroi SE, Perera SA, Pepper KW, Qi Q, Realini T, Richards JE, Ridker PM, Rimm E, Ritch R, Ritchie M, Schuman JS, Scott WK, Singh K, Sit AJ, Song YE, Tamimi RM, Topouzis F, Viswanathan AC, Verma SS, Vollrath D, Wang JJ, Weisschuh N, Wissinger B, Wollstein G, Wong TY, Yaspan BL, Zack DJ, Zhang K, Study ENE, Weinreb RN, Pericak-Vance MA, Small K, Hammond CJ, Aung T, Liu Y, Vithana EN, MacGregor S, Craig JE, Kraft P, Howell G, Hauser MA, Pasquale LR, Haines JL, Wiggs JL. Genome-wide association analysis identifies TXNRD2, ATXN2 and FOXC1 as susceptibility loci for primary open-angle glaucoma. Nat Genet 2016; 48:189-94. [PMID: 26752265 PMCID: PMC4731307 DOI: 10.1038/ng.3482] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022]
Abstract
Primary open angle glaucoma (POAG) is a leading cause of blindness world-wide. To identify new susceptibility loci, we meta-analyzed GWAS results from 8 independent studies from the United States (3,853 cases and 33,480 controls) and investigated the most significant SNPs in two Australian studies (1,252 cases and 2,592 controls), 3 European studies (875 cases and 4,107 controls) and a Singaporean Chinese study (1,037 cases and 2,543 controls). A meta-analysis of top SNPs identified three novel loci: rs35934224[T] within TXNRD2 (odds ratio (OR) = 0.78, P = 4.05×10−11 encoding a mitochondrial protein required for redox homeostasis; rs7137828[T] within ATXN2 (OR = 1.17, P = 8.73×10−10), and rs2745572[A] upstream of FOXC1 (OR = 1.17, P = 1.76×10−10). Using RT-PCR and immunohistochemistry, we show TXNRD2 and ATXN2 expression in retinal ganglion cells and the optic nerve head. These results identify new pathways underlying POAG susceptibility and suggest novel targets for preventative therapies.
Collapse
Affiliation(s)
- Jessica N Cooke Bailey
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Stephanie J Loomis
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Jae H Kang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - R Rand Allingham
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Chiea Chuen Khor
- Division of Human Genetics, Genome Institute of Singapore, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert P Igo
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Pirro G Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Craig A Glastonbury
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Allison Ashley-Koch
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Murray Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Andrew A Brown
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Donald L Budenz
- Department of Ophthalmology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Alfonso Buil
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Ching-Yu Cheng
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.,Eye Academic Clinical Program, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Hyon Choi
- Section of Rheumatology and Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Massachusetts, USA
| | - William G Christen
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gary Curhan
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John H Fingert
- Department of Ophthalmology, University of Iowa, College of Medicine, Iowa City, Iowa, USA.,Department of Anatomy and Cell Biology, University of Iowa, College of Medicine, Iowa City, Iowa, USA
| | - Paul J Foster
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital, London, UK.,Department of Ophthalmology, University College London, London, UK
| | - Charles Fuchs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Terry Gaasterland
- Scripps Genome Center, University of California at San Diego, San Diego, California, USA
| | - Alex W Hewitt
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia.,Department of Ophthalmology, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Frank Hu
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
| | - David J Hunter
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.,Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Anthony P Khawaja
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Zheng Li
- Division of Human Genetics, Genome Institute of Singapore, Singapore
| | - Paul R Lichter
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - David A Mackey
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Peter McGuffin
- Medical Research Council Social Genetic and Developmental Psychiatry Research Centre, Institute of Psychiatry, King's College London, London, UK
| | - Paul Mitchell
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Sayoko E Moroi
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Shamira A Perera
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.,Duke-National University of Singapore Graduate Medical School, Singapore
| | | | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Tony Realini
- Department of Ophthalmology, West Virginia University Eye Institute, Morgantown, West Virginia, USA
| | - Julia E Richards
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Rimm
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Robert Ritch
- Einhorn Clinical Research Center, Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Marylyn Ritchie
- Center for Systems Genomics, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Joel S Schuman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William K Scott
- Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kuldev Singh
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Arthur J Sit
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yeunjoo E Song
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Fotis Topouzis
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Ananth C Viswanathan
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital, London, UK
| | - Shefali Setia Verma
- Center for Systems Genomics, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jie Jin Wang
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Nicole Weisschuh
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Gadi Wollstein
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tien Y Wong
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | | | - Donald J Zack
- Wilmer Eye Institute, Johns Hopkins University Hospital, Baltimore, Maryland, USA
| | - Kang Zhang
- Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, San Diego, California, USA
| | - Epic-Norfolk Eye Study
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia, USA
| | | | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, San Diego, California, USA
| | - Margaret A Pericak-Vance
- Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kerrin Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.,Eye Academic Clinical Program, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia, USA.,James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Eranga N Vithana
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.,Eye Academic Clinical Program, Duke-National University of Singapore Graduate Medical School, Singapore
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.,Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts, USA
| | | | - Michael A Hauser
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| |
Collapse
|
131
|
Grotenhuis AJ, Dudek AM, Verhaegh GW, Aben KK, Witjes JA, Kiemeney LA, Vermeulen SH. Independent Replication of Published Germline Polymorphisms Associated with Urinary Bladder Cancer Prognosis and Treatment Response. Bladder Cancer 2016; 2:77-89. [PMID: 27376129 PMCID: PMC4927992 DOI: 10.3233/blc-150027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Many studies investigated the prognostic or predictive relevance of single nucleotide polymorphisms (SNPs) in biologically plausible genes in urinary bladder cancer (UBC) patients. Most published SNP associations have never been replicated in independent patient series. OBJECTIVE To independently replicate all previously reported associations between germline SNPs and disease prognosis or treatment response in UBC. METHODS A Pubmed search was performed to identify studies published by July 1, 2014 reporting on germline SNP associations with UBC prognosis or treatment response. For the replication series, consisting of 1,284 non-muscle-invasive bladder cancer (NMIBC) and 275 muscle-invasive or metastatic bladder cancer (MIBC) patients recruited through the Netherlands Cancer Registry, detailed clinical data were retrieved from medical charts. Patients were genotyped using a genome-wide SNP array. SNP association with recurrence-free, progression-free, and overall survival (OS) within specific patient and treatment strata was tested using Cox regression analyses. RESULTS For only six of the 114 evaluated SNPs, the association with either UBC prognosis or treatment response was replicated at the p < 0.05 level: rs1799793 (ERCC2) and rs187238 (IL18) for BCG recurrence; rs6678136 (RGS4) and rs11585883 (RGS5) for NMIBC progression; rs12035879 (RGS5) and rs2075786 (TERT) for MIBC OS. CONCLUSIONS Non-replicated genetic associations in the literature require cautious interpretation. This single replication does not provide definitive proof of association for the six SNPs, and non-replication of other SNPs may result from population-specific effects or the retrospective patient enrollment.
Collapse
Affiliation(s)
- Anne J Grotenhuis
- Radboud University Medical Center, Radboud Institute for Health Sciences , Nijmegen, The Netherlands
| | - Aleksandra M Dudek
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands
| | - Gerald W Verhaegh
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands
| | - Katja K Aben
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands; Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
| | - J Alfred Witjes
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands
| | - Lambertus A Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences , Nijmegen, The Netherlands
| | - Sita H Vermeulen
- Radboud University Medical Center, Radboud Institute for Health Sciences , Nijmegen, The Netherlands
| |
Collapse
|
132
|
Hu Y, Li H, Lu L, Manichaikul A, Zhu J, Chen YDI, Sun L, Liang S, Siscovick DS, Steffen LM, Tsai MY, Rich SS, Lemaitre RN, Lin X. Genome-wide meta-analyses identify novel loci associated with n-3 and n-6 polyunsaturated fatty acid levels in Chinese and European-ancestry populations. Hum Mol Genet 2016; 25:1215-24. [PMID: 26744325 DOI: 10.1093/hmg/ddw002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/05/2016] [Indexed: 11/12/2022] Open
Abstract
Epidemiological studies suggest that levels of n-3 and n-6 long-chain polyunsaturated fatty acids are associated with risk of cardio-metabolic outcomes across different ethnic groups. Recent genome-wide association studies in populations of European ancestry have identified several loci associated with plasma and/or erythrocyte polyunsaturated fatty acids. To identify additional novel loci, we carried out a genome-wide association study in two population-based cohorts consisting of 3521 Chinese participants, followed by a trans-ethnic meta-analysis with meta-analysis results from 8962 participants of European ancestry. Four novel loci (MYB, AGPAT4, DGAT2 and PPT2) reached genome-wide significance in the trans-ethnic meta-analysis (log10(Bayes Factor) ≥ 6). Of them, associations of MYB and AGPAT4 with docosatetraenoic acid (log10(Bayes Factor) = 11.5 and 8.69, respectively) also reached genome-wide significance in the Chinese-specific genome-wide association analyses (P = 4.15 × 10(-14) and 4.30 × 10(-12), respectively), while associations of DGAT2 with gamma-linolenic acid (log10(Bayes Factor) = 6.16) and of PPT2 with docosapentaenoic acid (log10(Bayes Factor) = 6.24) were nominally significant in both Chinese- and European-specific genome-wide association analyses (P ≤ 0.003). We also confirmed previously reported loci including FADS1, NTAN1, NRBF2, ELOVL2 and GCKR. Different effect sizes in FADS1 and independent association signals in ELOVL2 were observed. These results provide novel insight into the genetic background of polyunsaturated fatty acids and their differences between Chinese and European populations.
Collapse
Affiliation(s)
- Yao Hu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ling Lu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ani Manichaikul
- Center for Public Health Genomics and Department of Public Health Sciences, Biostatistics Section, University of Virginia, Charlottesville, VA, USA
| | - Jingwen Zhu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Liang Sun
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Shuang Liang
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - David S Siscovick
- Cardiovascular Health Research Unit, Department of Medicine and Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA, New York Academy of Medicine, New York, NY, USA and
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, People's Republic of China,
| |
Collapse
|
133
|
Delahaye-Sourdeix M, Urayama KY, Gaborieau V, Veenstra R, Foll M, Chabrier A, Benavente Y, Nieters A, Becker N, Foretova L, Maynadié M, Staines A, Smedby KE, Glimelius I, Lightfoot T, Cocco P, Galan P, Vatten LJ, Duell EJ, Kiemeney L, Roman E, de Sanjosé S, Lathrop M, Melbye M, Brennan P, Diepstra A, van den Berg A, Hjalgrim H, Jarrett RF, McKay JD. A Novel Risk Locus at 6p21.3 for Epstein-Barr Virus-Positive Hodgkin Lymphoma. Cancer Epidemiol Biomarkers Prev 2015; 24:1838-43. [PMID: 26404960 DOI: 10.1158/1055-9965.epi-15-0534] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/26/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND A proportion of the genetic variants involved in susceptibility to Hodgkin lymphoma differ by the tumor's Epstein-Barr virus (EBV) status, particularly within the MHC region. METHODS We have conducted an SNP imputation study of the MHC region, considering tumor EBV status in 1,200 classical Hodgkin lymphoma (cHL) cases and 5,726 control subjects of European origin. Notable findings were genotyped in an independent study population of 468 cHL cases and 551 controls. RESULTS We identified and subsequently replicated a novel association between a common genetic variant rs6457715 and cHL. Although strongly associated with EBV-positive cHL [OR, 2.33; 95% confidence interval (CI), 1.83-2.97; P = 7 × 10(-12)], there was little evidence for association between rs6457715 and the EBV-negative subgroup of cHL (OR, 1.06; 95% CI, 0.92-1.21), indicating that this association was specific to the EBV-positive subgroup (Phet < P = 10(-8)). Furthermore, the association was limited to EBV-positive cHL subgroups within mixed cell (MCHL) and nodular sclerosis subtypes (NSHL), suggesting that the association is independent of histologic subtype of cHL. CONCLUSIONS rs6457715, located near the HLA-DPB1 gene, is associated with EBV-positive cHL and suggests this region as a novel susceptibility locus for cHL. IMPACT This expands the number of genetic variants that are associated with cHL and provides additional evidence for a critical and specific role of EBV in the etiology of this disease.
Collapse
Affiliation(s)
| | - Kevin Y Urayama
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Rianne Veenstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Matthieu Foll
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Amelie Chabrier
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Yolanda Benavente
- Unit of Infections and Cancer, Cancer Epidemiology Research Programme, IDIBELL, Catalan Institute of Oncology, Barcelona, Spain. CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Alexandra Nieters
- Centre of Chronic Immunodeficiency Freiburg, University Medical Centre Freiburg, Freiburg, Germany
| | - Nikolaus Becker
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Marc Maynadié
- Registry of Hematological Malignancies, University of Burgundy, University Hospital of Dijon, Dijon, France
| | - Anthony Staines
- School of Nursing, Dublin City University, Glasnevin, Dublin, Ireland
| | - Karin Ekstrom Smedby
- Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ingrid Glimelius
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden. Department of Radiology, Oncology, and Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - Tracy Lightfoot
- Epidemiology and Genetics Unit, Department of Health Sciences, University of York, York, United Kingdom
| | - Pierluigi Cocco
- Department of Public Health, Occupational Health Section, University of Cagliari, Cagliari, Italy
| | - Pilar Galan
- Sorbonne Paris Cité Epidemiology and Biostatistics Research Center (CRESS), Nutritional Epidemiology Research Team (EREN), Inserm U1153, Inra U1125, Cnam, University Paris 13, University Paris 5, University Paris 7, Bobigny, France
| | - Lars J Vatten
- Norwegian University of Science and Technology, Trondheim, Norway
| | - Eric J Duell
- Unit of Infections and Cancer, Cancer Epidemiology Research Programme, IDIBELL, Catalan Institute of Oncology, Barcelona, Spain
| | - Lambertus Kiemeney
- Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, the Netherlands
| | - Eve Roman
- Epidemiology and Genetics Unit, Department of Health Sciences, University of York, York, United Kingdom
| | - Silvia de Sanjosé
- Unit of Infections and Cancer, Cancer Epidemiology Research Programme, IDIBELL, Catalan Institute of Oncology, Barcelona, Spain. CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Mark Lathrop
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ruth F Jarrett
- MRC University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - James D McKay
- International Agency for Research on Cancer (IARC), Lyon, France.
| |
Collapse
|
134
|
Binder M, Shui IM, Wilson KM, Penney KL, Mucci LA, Kibel AS. Calcium intake, polymorphisms of the calcium-sensing receptor, and recurrent/aggressive prostate cancer. Cancer Causes Control 2015; 26:1751-9. [PMID: 26407952 PMCID: PMC4633306 DOI: 10.1007/s10552-015-0668-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 09/11/2015] [Indexed: 11/28/2022]
Abstract
PURPOSE To assess whether calcium intake and common genetic variants of the calcium-sensing receptor (CASR) are associated with either aggressive prostate cancer (PCa) or disease recurrence after prostatectomy. METHODS Calcium intake at diagnosis was assessed, and 65 common single-nucleotide polymorphisms (SNPs) in CASR were genotyped in 886 prostatectomy patients. We investigated the association between calcium intake and CASR variants with both PCa recurrence and aggressiveness (defined as Gleason score ≥4 + 3, stage ≥pT3, or nodal-positive disease). RESULTS A total of 285 men had aggressive disease and 91 experienced recurrence. A U-shaped relationship between calcium intake and both disease recurrence and aggressiveness was observed. Compared to the middle quintile, the HR for disease recurrence was 3.07 (95% CI 1.41-6.69) for the lowest quintile and 3.21 (95% CI 1.47-7.00) and 2.97 (95% CI 1.37-6.45) for the two upper quintiles, respectively. Compared to the middle quintile, the OR for aggressive disease was 1.80 (95% CI 1.11-2.91) for the lowest quintile and 1.75 (95% CI 1.08-2.85) for the highest quintile of calcium intake. The main effects of CASR variants were not associated with PCa recurrence or aggressiveness. In the subgroup of patients with moderate calcium intake, 31 SNPs in four distinct blocks of high linkage disequilibrium were associated with PCa recurrence. CONCLUSIONS We observed a protective effect of moderate calcium intake for PCa aggressiveness and recurrence. While CASR variants were not associated with these outcomes in the entire cohort, they may be associated with disease recurrence in men with moderate calcium intakes.
Collapse
Affiliation(s)
- Moritz Binder
- Master of Public Health Program, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Irene M Shui
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Kathryn M Wilson
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Brigham & Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02215, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Brigham & Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02215, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Brigham & Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02215, USA
| | - Adam S Kibel
- Division of Urologic Surgery, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
135
|
Dong J, Yang J, Tranah G, Franceschini N, Parimi N, Alkorta-Aranburu G, Xu Z, Alonso A, Cummings SR, Fornage M, Huang X, Kritchevsky S, Liu Y, London S, Niu L, Wilson RS, De Jager PL, Yu L, Singleton AB, Harris T, Mosley TH, Pinto JM, Bennett DA, Chen H. Genome-wide Meta-analysis on the Sense of Smell Among US Older Adults. Medicine (Baltimore) 2015; 94:e1892. [PMID: 26632684 PMCID: PMC5058953 DOI: 10.1097/md.0000000000001892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/04/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022] Open
Abstract
Olfactory dysfunction is common among older adults and affects their safety, nutrition, quality of life, and mortality. More importantly, the decreased sense of smell is an early symptom of neurodegenerative diseases such as Parkinson disease (PD) and Alzheimer disease. However, the genetic determinants for the sense of smell have been poorly investigated. We here performed the first genome-wide meta-analysis on the sense of smell among 6252 US older adults of European descent from the Atherosclerosis Risk in Communities (ARIC) study, the Health, Aging, and Body Composition (Health ABC) study, and the Religious Orders Study and the Rush Memory and Aging Project (ROS/MAP). Genome-wide association study analysis was performed first by individual cohorts and then meta-analyzed using fixed-effect models with inverse variance weights. Although no SNPs reached genome-wide statistical significance, we identified 13 loci with suggestive evidence for an association with the sense of smell (Pmeta < 1 × 10). Of these, 2 SNPs at chromosome 17q21.31 (rs199443 in NSF, P = 3.02 × 10; and rs2732614 in KIAA1267-LRRC37A, P = 6.65 × 10) exhibited cis effects on the expression of microtubule-associated protein tau (MAPT, 17q21.31) in 447 frontal-cortex samples obtained postmortem and profiled by RNA-seq (P < 1 × 10). Gene-based and pathway-enrichment analyses further implicated MAPT in regulating the sense of smell in older adults. Similar results were obtained after excluding participants who reported a physician-diagnosed PD or use of PD medications. In conclusion, we provide preliminary evidence that the MAPT locus may play a role in regulating the sense of smell in older adults and therefore offer a potential genetic link between poor sense of smell and major neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Dong
- From the Epidemiology Branch (JD, ZX, SL, HC) and Biostatistics Branch (LN), National Institute of Environmental Health Sciences, Research Triangle Park, NC; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (JY, RSW, LY, DAB); California Pacific Medical Center Research Institute, San Francisco, CA (GT, NP, SC); Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC (NF); Department of Human Genetics, University of Chicago, Chicago, IL (GA-A); Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN (AA); Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX (MF); Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, PA (XH); Sticht Center on Aging (SK) and Division of Public Health Sciences (YL), Wake Forest School of Medicine, Winston-Salem, NC; Program in Translational Neuro Psychiatric Genomics, Departments of Neurology and Psychiatry, Institute for the Neurosciences, Brigham and Women's Hospital; Harvard Medical School; Program in Medical and Population Genetics, Broad Institute, Boston, MA (PLD); Laboratory of Neurogenetics (ABS) and Laboratory of Epidemiology, Demography, and Biometry (TH), National Institute on Aging, Bethesda, MD; Division of Geriatrics, Department of Medicine, University of Mississippi Medical Center, Jackson, MS (THM); Section of Otolaryngology-Head and Neck Surgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL (JMP); Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA (JY, LY, DAB); and Departments of Neurological Sciences and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA (RSW)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Coleman JRI, Euesden J, Patel H, Folarin AA, Newhouse S, Breen G. Quality control, imputation and analysis of genome-wide genotyping data from the Illumina HumanCoreExome microarray. Brief Funct Genomics 2015; 15:298-304. [PMID: 26443613 DOI: 10.1093/bfgp/elv037] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The decreasing cost of performing genome-wide association studies has made genomics widely accessible. However, there is a paucity of guidance for best practice in conducting such analyses. For the results of a study to be valid and replicable, multiple biases must be addressed in the course of data preparation and analysis. In addition, standardizing methods across small, independent studies would increase comparability and the potential for effective meta-analysis. This article provides a discussion of important aspects of quality control, imputation and analysis of genome-wide data from a low-coverage microarray, as well as a straight-forward guide to performing a genome-wide association study. A detailed protocol is provided online, with example scripts available at https://github.com/JoniColeman/gwas_scripts.
Collapse
|
137
|
Hancock DB, Reginsson GW, Gaddis NC, Chen X, Saccone NL, Lutz SM, Qaiser B, Sherva R, Steinberg S, Zink F, Stacey SN, Glasheen C, Chen J, Gu F, Frederiksen BN, Loukola A, Gudbjartsson DF, Brüske I, Landi MT, Bickeböller H, Madden P, Farrer L, Kaprio J, Kranzler HR, Gelernter J, Baker TB, Kraft P, Amos CI, Caporaso NE, Hokanson JE, Bierut LJ, Thorgeirsson TE, Johnson EO, Stefansson K. Genome-wide meta-analysis reveals common splice site acceptor variant in CHRNA4 associated with nicotine dependence. Transl Psychiatry 2015; 5:e651. [PMID: 26440539 PMCID: PMC4930126 DOI: 10.1038/tp.2015.149] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/19/2015] [Indexed: 01/04/2023] Open
Abstract
We conducted a 1000 Genomes-imputed genome-wide association study (GWAS) meta-analysis for nicotine dependence, defined by the Fagerström Test for Nicotine Dependence in 17 074 ever smokers from five European-ancestry samples. We followed up novel variants in 7469 ever smokers from five independent European-ancestry samples. We identified genome-wide significant association in the alpha-4 nicotinic receptor subunit (CHRNA4) gene on chromosome 20q13: lowest P=8.0 × 10(-9) across all the samples for rs2273500-C (frequency=0.15; odds ratio=1.12 and 95% confidence interval=1.08-1.17 for severe vs mild dependence). rs2273500-C, a splice site acceptor variant resulting in an alternate CHRNA4 transcript predicted to be targeted for nonsense-mediated decay, was associated with decreased CHRNA4 expression in physiologically normal human brains (lowest P=7.3 × 10(-4)). Importantly, rs2273500-C was associated with increased lung cancer risk (N=28 998, odds ratio=1.06 and 95% confidence interval=1.00-1.12), likely through its effect on smoking, as rs2273500-C was no longer associated with lung cancer after adjustment for smoking. Using criteria for smoking behavior that encompass more than the single 'cigarettes per day' item, we identified a common CHRNA4 variant with important regulatory properties that contributes to nicotine dependence and smoking-related consequences.
Collapse
Affiliation(s)
- D B Hancock
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Research Division, Research Triangle Institute International, Research Triangle Park, NC, USA,Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Research Division, Research Triangle Institute International, 3040 East Cornwallis Road, P.O. Box 12194, Research Triangle Park, NC 27709, USA. E-mail:
| | | | - N C Gaddis
- Research Computing Division, Research Triangle Institute International, Research Triangle Park, NC, USA
| | - X Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA,Nevada Institute of Personalized Medicine and Department of Psychology, University of Nevada, Las Vegas, NV, USA
| | - N L Saccone
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - S M Lutz
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - B Qaiser
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Sherva
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | | | - F Zink
- deCODE Genetics/Amgen, Reykjavik, Iceland
| | - S N Stacey
- deCODE Genetics/Amgen, Reykjavik, Iceland
| | - C Glasheen
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Research Division, Research Triangle Institute International, Research Triangle Park, NC, USA
| | - J Chen
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - F Gu
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
| | | | - A Loukola
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - I Brüske
- Institute of Epidemiology I, German Research Center for Environmental Health, Neuherberg, Germany
| | - M T Landi
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
| | - H Bickeböller
- Department of Genetic Epidemiology, University of Göttingen—Georg-August University Göttingen, Göttingen, Germany
| | - P Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - L Farrer
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA,Department of Neurology, Boston University School of Medicine, Boston, MA, USA,Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA,Department of Genetics and Genomics, Boston University School of Medicine, Boston, MA, USA,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - J Kaprio
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland,National Institute for Health and Welfare, Helsinki, Finland,Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - H R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA,VISN 4 Mental Illness Research, Education and Clinical Center, Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - J Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Department of Genetics, Yale University School of Medicine, New Haven, CT, USA,Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA,VA CT Healthcare Center, Department of Psychiatry, West Haven, CT, USA
| | - T B Baker
- Center for Tobacco Research and Intervention, University of Wisconsin, Madison, WI, USA
| | - P Kraft
- Department of Epidemiology, Harvard University School of Public Health, Boston, MA, USA,Department of Biostatistics, Harvard University School of Public Health, Boston, MA, USA
| | - C I Amos
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanoven, NH, USA
| | - N E Caporaso
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, USA
| | - J E Hokanson
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - L J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - E O Johnson
- Fellow Program and Behavioral Health and Criminal Justice Research Division, Research Triangle Institute International, Research Triangle Park, NC, USA
| | - K Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
138
|
Winkler TW, Justice AE, Graff M, Barata L, Feitosa MF, Chu S, Czajkowski J, Esko T, Fall T, Kilpeläinen TO, Lu Y, Mägi R, Mihailov E, Pers TH, Rüeger S, Teumer A, Ehret GB, Ferreira T, Heard-Costa NL, Karjalainen J, Lagou V, Mahajan A, Neinast MD, Prokopenko I, Simino J, Teslovich TM, Jansen R, Westra HJ, White CC, Absher D, Ahluwalia TS, Ahmad S, Albrecht E, Alves AC, Bragg-Gresham JL, de Craen AJM, Bis JC, Bonnefond A, Boucher G, Cadby G, Cheng YC, Chiang CWK, Delgado G, Demirkan A, Dueker N, Eklund N, Eiriksdottir G, Eriksson J, Feenstra B, Fischer K, Frau F, Galesloot TE, Geller F, Goel A, Gorski M, Grammer TB, Gustafsson S, Haitjema S, Hottenga JJ, Huffman JE, Jackson AU, Jacobs KB, Johansson Å, Kaakinen M, Kleber ME, Lahti J, Leach IM, Lehne B, Liu Y, Lo KS, Lorentzon M, Luan J, Madden PAF, Mangino M, McKnight B, Medina-Gomez C, Monda KL, Montasser ME, Müller G, Müller-Nurasyid M, Nolte IM, Panoutsopoulou K, Pascoe L, Paternoster L, Rayner NW, Renström F, Rizzi F, Rose LM, Ryan KA, Salo P, Sanna S, Scharnagl H, Shi J, Smith AV, Southam L, Stančáková A, Steinthorsdottir V, Strawbridge RJ, Sung YJ, Tachmazidou I, et alWinkler TW, Justice AE, Graff M, Barata L, Feitosa MF, Chu S, Czajkowski J, Esko T, Fall T, Kilpeläinen TO, Lu Y, Mägi R, Mihailov E, Pers TH, Rüeger S, Teumer A, Ehret GB, Ferreira T, Heard-Costa NL, Karjalainen J, Lagou V, Mahajan A, Neinast MD, Prokopenko I, Simino J, Teslovich TM, Jansen R, Westra HJ, White CC, Absher D, Ahluwalia TS, Ahmad S, Albrecht E, Alves AC, Bragg-Gresham JL, de Craen AJM, Bis JC, Bonnefond A, Boucher G, Cadby G, Cheng YC, Chiang CWK, Delgado G, Demirkan A, Dueker N, Eklund N, Eiriksdottir G, Eriksson J, Feenstra B, Fischer K, Frau F, Galesloot TE, Geller F, Goel A, Gorski M, Grammer TB, Gustafsson S, Haitjema S, Hottenga JJ, Huffman JE, Jackson AU, Jacobs KB, Johansson Å, Kaakinen M, Kleber ME, Lahti J, Leach IM, Lehne B, Liu Y, Lo KS, Lorentzon M, Luan J, Madden PAF, Mangino M, McKnight B, Medina-Gomez C, Monda KL, Montasser ME, Müller G, Müller-Nurasyid M, Nolte IM, Panoutsopoulou K, Pascoe L, Paternoster L, Rayner NW, Renström F, Rizzi F, Rose LM, Ryan KA, Salo P, Sanna S, Scharnagl H, Shi J, Smith AV, Southam L, Stančáková A, Steinthorsdottir V, Strawbridge RJ, Sung YJ, Tachmazidou I, Tanaka T, Thorleifsson G, Trompet S, Pervjakova N, Tyrer JP, Vandenput L, van der Laan SW, van der Velde N, van Setten J, van Vliet-Ostaptchouk JV, Verweij N, Vlachopoulou E, Waite LL, Wang SR, Wang Z, Wild SH, Willenborg C, Wilson JF, Wong A, Yang J, Yengo L, Yerges-Armstrong LM, Yu L, Zhang W, Zhao JH, Andersson EA, Bakker SJL, Baldassarre D, Banasik K, Barcella M, Barlassina C, Bellis C, Benaglio P, Blangero J, Blüher M, Bonnet F, Bonnycastle LL, Boyd HA, Bruinenberg M, Buchman AS, Campbell H, Chen YDI, Chines PS, Claudi-Boehm S, Cole J, Collins FS, de Geus EJC, de Groot LCPGM, Dimitriou M, Duan J, Enroth S, Eury E, Farmaki AE, Forouhi NG, Friedrich N, Gejman PV, Gigante B, Glorioso N, Go AS, Gottesman O, Gräßler J, Grallert H, Grarup N, Gu YM, Broer L, Ham AC, Hansen T, Harris TB, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Heath AC, Henders AK, Hernandez D, Hillege H, Holmen O, Hovingh KG, Hui J, Husemoen LL, Hutri-Kähönen N, Hysi PG, Illig T, De Jager PL, Jalilzadeh S, Jørgensen T, Jukema JW, Juonala M, Kanoni S, Karaleftheri M, Khaw KT, Kinnunen L, Kittner SJ, Koenig W, Kolcic I, Kovacs P, Krarup NT, Kratzer W, Krüger J, Kuh D, Kumari M, Kyriakou T, Langenberg C, Lannfelt L, Lanzani C, Lotay V, Launer LJ, Leander K, Lindström J, Linneberg A, Liu YP, Lobbens S, Luben R, Lyssenko V, Männistö S, Magnusson PK, McArdle WL, Menni C, Merger S, Milani L, Montgomery GW, Morris AP, Narisu N, Nelis M, Ong KK, Palotie A, Pérusse L, Pichler I, Pilia MG, Pouta A, Rheinberger M, Ribel-Madsen R, Richards M, Rice KM, Rice TK, Rivolta C, Salomaa V, Sanders AR, Sarzynski MA, Scholtens S, Scott RA, Scott WR, Sebert S, Sengupta S, Sennblad B, Seufferlein T, Silveira A, Slagboom PE, Smit JH, Sparsø TH, Stirrups K, Stolk RP, Stringham HM, Swertz MA, Swift AJ, Syvänen AC, Tan ST, Thorand B, Tönjes A, Tremblay A, Tsafantakis E, van der Most PJ, Völker U, Vohl MC, Vonk JM, Waldenberger M, Walker RW, Wennauer R, Widén E, Willemsen G, Wilsgaard T, Wright AF, Zillikens MC, van Dijk SC, van Schoor NM, Asselbergs FW, de Bakker PIW, Beckmann JS, Beilby J, Bennett DA, Bergman RN, Bergmann S, Böger CA, Boehm BO, Boerwinkle E, Boomsma DI, Bornstein SR, Bottinger EP, Bouchard C, Chambers JC, Chanock SJ, Chasman DI, Cucca F, Cusi D, Dedoussis G, Erdmann J, Eriksson JG, Evans DA, de Faire U, Farrall M, Ferrucci L, Ford I, Franke L, Franks PW, Froguel P, Gansevoort RT, Gieger C, Grönberg H, Gudnason V, Gyllensten U, Hall P, Hamsten A, van der Harst P, Hayward C, Heliövaara M, Hengstenberg C, Hicks AA, Hingorani A, Hofman A, Hu F, Huikuri HV, Hveem K, James AL, Jordan JM, Jula A, Kähönen M, Kajantie E, Kathiresan S, Kiemeney LALM, Kivimaki M, Knekt PB, Koistinen HA, Kooner JS, Koskinen S, Kuusisto J, Maerz W, Martin NG, Laakso M, Lakka TA, Lehtimäki T, Lettre G, Levinson DF, Lind L, Lokki ML, Mäntyselkä P, Melbye M, Metspalu A, Mitchell BD, Moll FL, Murray JC, Musk AW, Nieminen MS, Njølstad I, Ohlsson C, Oldehinkel AJ, Oostra BA, Palmer LJ, Pankow JS, Pasterkamp G, Pedersen NL, Pedersen O, Penninx BW, Perola M, Peters A, Polašek O, Pramstaller PP, Psaty BM, Qi L, Quertermous T, Raitakari OT, Rankinen T, Rauramaa R, Ridker PM, Rioux JD, Rivadeneira F, Rotter JI, Rudan I, den Ruijter HM, Saltevo J, Sattar N, Schunkert H, Schwarz PEH, Shuldiner AR, Sinisalo J, Snieder H, Sørensen TIA, Spector TD, Staessen JA, Stefania B, Thorsteinsdottir U, Stumvoll M, Tardif JC, Tremoli E, Tuomilehto J, Uitterlinden AG, Uusitupa M, Verbeek ALM, Vermeulen SH, Viikari JS, Vitart V, Völzke H, Vollenweider P, Waeber G, Walker M, Wallaschofski H, Wareham NJ, Watkins H, Zeggini E, Chakravarti A, Clegg DJ, Cupples LA, Gordon-Larsen P, Jaquish CE, Rao DC, Abecasis GR, Assimes TL, Barroso I, Berndt SI, Boehnke M, Deloukas P, Fox CS, Groop LC, Hunter DJ, Ingelsson E, Kaplan RC, McCarthy MI, Mohlke KL, O'Connell JR, Schlessinger D, Strachan DP, Stefansson K, van Duijn CM, Hirschhorn JN, Lindgren CM, Heid IM, North KE, Borecki IB, Kutalik Z, Loos RJF. The Influence of Age and Sex on Genetic Associations with Adult Body Size and Shape: A Large-Scale Genome-Wide Interaction Study. PLoS Genet 2015; 11:e1005378. [PMID: 26426971 PMCID: PMC4591371 DOI: 10.1371/journal.pgen.1005378] [Show More Authors] [Citation(s) in RCA: 275] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 06/22/2015] [Indexed: 01/11/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified more than 100 genetic variants contributing to BMI, a measure of body size, or waist-to-hip ratio (adjusted for BMI, WHRadjBMI), a measure of body shape. Body size and shape change as people grow older and these changes differ substantially between men and women. To systematically screen for age- and/or sex-specific effects of genetic variants on BMI and WHRadjBMI, we performed meta-analyses of 114 studies (up to 320,485 individuals of European descent) with genome-wide chip and/or Metabochip data by the Genetic Investigation of Anthropometric Traits (GIANT) Consortium. Each study tested the association of up to ~2.8M SNPs with BMI and WHRadjBMI in four strata (men ≤50y, men >50y, women ≤50y, women >50y) and summary statistics were combined in stratum-specific meta-analyses. We then screened for variants that showed age-specific effects (G x AGE), sex-specific effects (G x SEX) or age-specific effects that differed between men and women (G x AGE x SEX). For BMI, we identified 15 loci (11 previously established for main effects, four novel) that showed significant (FDR<5%) age-specific effects, of which 11 had larger effects in younger (<50y) than in older adults (≥50y). No sex-dependent effects were identified for BMI. For WHRadjBMI, we identified 44 loci (27 previously established for main effects, 17 novel) with sex-specific effects, of which 28 showed larger effects in women than in men, five showed larger effects in men than in women, and 11 showed opposite effects between sexes. No age-dependent effects were identified for WHRadjBMI. This is the first genome-wide interaction meta-analysis to report convincing evidence of age-dependent genetic effects on BMI. In addition, we confirm the sex-specificity of genetic effects on WHRadjBMI. These results may provide further insights into the biology that underlies weight change with age or the sexually dimorphism of body shape.
Collapse
Affiliation(s)
- Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University Regensburg, Regensburg, Germany
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Llilda Barata
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Su Chu
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jacek Czajkowski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Tõnu Esko
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts, United States of America
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tove Fall
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Reedik Mägi
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Tune H. Pers
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Sina Rüeger
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), Lausanne, Switzerland
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg B. Ehret
- Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Nancy L. Heard-Costa
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Juha Karjalainen
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Michael D. Neinast
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
- Hammersmith Hospital, London, United Kingdom
- Department of Genomics of Common Diseases, School of Public Health, Imperial College London, London, United Kingdom
| | - Jeannette Simino
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Tanya M. Teslovich
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Rick Jansen
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Harm-Jan Westra
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
| | - Charles C. White
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Tarunveer S. Ahluwalia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center A/S, Gentofte, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Shafqat Ahmad
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, 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, United Kingdom
| | - Jennifer L. Bragg-Gresham
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Amélie Bonnefond
- CNRS UMR 8199, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille 2, Lille, France
| | | | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Western Australia, Australia
| | - Yu-Ching Cheng
- VA Maryland Health Care System, Baltimore, Maryland, United States of America
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, United States of America
| | - Charleston W. K. Chiang
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Graciela Delgado
- Vth Department of Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Ayse Demirkan
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole Dueker
- Universiy of Maryland School of Medicine, Department of Epidemiology & Public Health, Baltimore, Maryland, United States of America
| | - Niina Eklund
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
- National Institute for Health and Welfare, Public Health Genomics Unit, Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | | | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Krista Fischer
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
| | - Francesca Frau
- Department of Health Sciences, University of Milan, Milan, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano, Italy
| | - Tessel E. Galesloot
- Radboud university medical center, Radboud Institute for Health Sciences, Department for Health Evidence, Nijmegen, The Netherlands
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anuj Goel
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovacular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University Regensburg, Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tanja B. Grammer
- Vth Department of Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Stefan Gustafsson
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Saskia Haitjema
- Experimental Cardiology and laboratory of clinical chemistry, UMCU, Utrecht, The Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Jennifer E. Huffman
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts, United States of America
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Anne U. Jackson
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin B. Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Core Genotyping Facility, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | - Åsa Johansson
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marika Kaakinen
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, United Kingdom
- Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Marcus E. Kleber
- Vth Department of Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Jari Lahti
- Folkhälsan Research Centre, Helsinki, Finland
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Irene Mateo Leach
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, Netherlands
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chaper Hill, North Carolina, United States of America
| | - Ken Sin Lo
- Montreal Heart Institute, Montréal, Québec, Canada
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Pamela A. F. Madden
- Washington University Medical School, St. Louis, Missouri, United States of America
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Program in Biostatistics and Biomathematics, Divison of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Carolina Medina-Gomez
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Keri L. Monda
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- The Center for Observational Research, Amgen Inc., Thousand Oaks, California, United States of America
| | - May E. Montasser
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Dept of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Gabriele Müller
- Center for Evidence Based Healthcare, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, Munich, Germany
| | - Ilja M. Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | | | - Laura Pascoe
- Institute of Cell & Molecular Biosciences, Newcastle University, Newcastle, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Nigel W. Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
| | - Federica Rizzi
- Department of Health Sciences, University of Milan, Milan, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano, Italy
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Kathy A. Ryan
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Dept of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Perttu Salo
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
- National Institute for Health and Welfare, Public Health Genomics Unit, Helsinki, Finland
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato, Italy
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Jianxin Shi
- National Cancer Institute, Bethesda, Maryland, United States of America
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Lorraine Southam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Rona J. Strawbridge
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ioanna Tachmazidou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | | | - Stella Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Natalia Pervjakova
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
- National Institute for Health and Welfare, Public Health Genomics Unit, Helsinki, Finland
- Institute of Cell and Molecular Biology, Department of Biotechnology, University of Tartu, Tartu, Estonia
- University of Helsinki, Helsinki, Finland
| | - Jonathan P. Tyrer
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sander W van der Laan
- Experimental Cardiology and laboratory of clinical chemistry, UMCU, Utrecht, The Netherlands
| | - Nathalie van der Velde
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Section of Geriatric Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jessica van Setten
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jana V. van Vliet-Ostaptchouk
- University of Groningen, University Medical Center Groningen, Department of Endocrinology, Groningen, The Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, Netherlands
| | - Efthymia Vlachopoulou
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Lindsay L. Waite
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Sophie R. Wang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, United States of America
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts, United States of America
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Core Genotyping Facility, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | - Sarah H. Wild
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Christina Willenborg
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany
| | - James F. Wilson
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, London, United Kingdom
| | - Jian Yang
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Loïc Yengo
- CNRS UMR 8199, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille 2, Lille, France
| | - Laura M. Yerges-Armstrong
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Dept of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Ealing Hospital NHS Trust, Middlesex, United Kingdom
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Ehm A. Andersson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephan J. L. Bakker
- University of Groningen, University Medical Center Groningen, Department of Medicine, Groningen, Netherlands
| | - Damiano Baldassarre
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
| | - Karina Banasik
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matteo Barcella
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Claire Bellis
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
- Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paola Benaglio
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - John Blangero
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | | | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Heather A. Boyd
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Marcel Bruinenberg
- University of Groningen, University Medical Center Groningen, The LifeLines Cohort Study, Groningen, The Netherlands
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Yii-Der Ida Chen
- Los Angeles BioMedical Resesarch Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Simone Claudi-Boehm
- Department of Internal Medicine I, Ulm University Medical Centre, Ulm, Germany
| | - John Cole
- VA Maryland Health Care System, Baltimore, Maryland, United States of America
- Universiy of Maryland School of Medicine, Department of Neurology, Baltimore, Maryland, United States of America
| | - Francis S. Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Eco J. C. de Geus
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Maria Dimitriou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- Department of Dietetics-Nutrition, Harokopio University, Athens, Greece
| | - Jubao Duan
- NorthShore University HealthSystem, Evanston, Illinois, United States of America
- University of Chicago, Chicago, Illinois, United States of America
| | - Stefan Enroth
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Elodie Eury
- CNRS UMR 8199, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille 2, Lille, France
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Nita G. Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Pablo V. Gejman
- NorthShore University HealthSystem, Evanston, Illinois, United States of America
- University of Chicago, Chicago, Illinois, United States of America
| | - Bruna Gigante
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari, Sassari, Italy
| | - Alan S. Go
- Kaiser Permanente, Division of Research, Oakland, California, United States of America
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jürgen Gräßler
- Department of Medicine III, Pathobiochemistry, University of Dresden, Dresden, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yu-Mei Gu
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Linda Broer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annelies C. Ham
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland, United States of America
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Catharina A. Hartman
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Andrew T. Hattersley
- Institue of Biomedical & Clinical Science, University of Exeter, Exeter, United Kingdom
| | - Andrew C. Heath
- Washington University Medical School, St. Louis, Missouri, United States of America
| | - Anjali K. Henders
- QIMR Bergofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hans Hillege
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, Netherlands
| | - Oddgeir Holmen
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kees G Hovingh
- Department Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jennie Hui
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia,Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
- School of Population Health, University of Western Australia, Nedlands, Western Australia, Australia
| | - Lise L. Husemoen
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
| | - Nina Hutri-Kähönen
- Department of Pediatrics, University of Tampere School of Medicine, Tampere, Finland
- Department of Pediatrics,Tampere University Hospital, Tampere, Finland
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
- Institute of Human Genetics, Hannover Medical School, Hanover, Germany
| | - Philip L. De Jager
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts,United States of America
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts,United States of America
| | - Shapour Jalilzadeh
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovacular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg, Aalborg, Denmark
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Markus Juonala
- Division of Medicine, Turku University Hospital, Turku, Finland
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Medicine, University of Turku, Turku, Finland
| | - Stavroula Kanoni
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Kay Tee Khaw
- Clinical Gerontology Unit, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Leena Kinnunen
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Steven J. Kittner
- VA Maryland Health Care System, Baltimore, Maryland, United States of America
- Universiy of Maryland School of Medicine, Department of Neurology, Baltimore, Maryland, United States of America
| | - Wolfgang Koenig
- Department of Internal Medicine II—Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Nikolaj T. Krarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wolfgang Kratzer
- Department of Internal Medicine I, Ulm University Medical Centre, Ulm, Germany
| | - Janine Krüger
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Diana Kuh
- MRC Unit for Lifelong Health & Ageing at UCL, London, United Kingdom
| | - Meena Kumari
- Department of Epidemiology and Public Health, UCL, London, United Kingdom
| | - Theodosios Kyriakou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovacular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- Department of Epidemiology and Public Health, UCL, London, United Kingdom
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Chiara Lanzani
- Chair of Nephrology, Università Vita Salute San Raffaele, Segrate (Milan), Italy
- Genomics of Renal Disease and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Segrate (Milan), Italy
| | - Vaneet Lotay
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Lenore J. Launer
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Karin Leander
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jaana Lindström
- Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
| | - Yan-Ping Liu
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Stéphane Lobbens
- CNRS UMR 8199, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille 2, Lille, France
| | - Robert Luben
- Strangeways Research Laboratory Wort's Causeway, Cambridge, United Kingdom
| | - Valeriya Lyssenko
- Steno Diabetes Center A/S, Gentofte, Denmark
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, Malmö, Sweden
| | - Satu Männistö
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
| | - Patrik K. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Sigrun Merger
- Department of Internal Medicine I, Ulm University Medical Centre, Ulm, Germany
| | - Lili Milani
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
| | | | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Mari Nelis
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
| | - Ken K. Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- MRC Unit for Lifelong Health & Ageing at UCL, London, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- Massachusetts General Hospital, Center for Human Genetic Research, Psychiatric and Neurodevelopmental Genetics Unit, Boston, Massachusetts, United States of America
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| | - Irene Pichler
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy, Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Maria G. Pilia
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato, Italy
| | - Anneli Pouta
- Department of Children, Young People and Families, National Institute for Health and Welfare, Helsinki, Finland
- Department of Obstetrics and Gynecology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Myriam Rheinberger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Rasmus Ribel-Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marcus Richards
- MRC Unit for Lifelong Health & Ageing at UCL, London, United Kingdom
| | - Kenneth M. Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Carlo Rivolta
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
| | - Alan R. Sanders
- NorthShore University HealthSystem, Evanston, Illinois, United States of America
- University of Chicago, Chicago, Illinois, United States of America
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America
| | - Salome Scholtens
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Robert A. Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - William R. Scott
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Ealing Hospital NHS Trust, Middlesex, United Kingdom
| | - Sylvain Sebert
- Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Sebanti Sengupta
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bengt Sennblad
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Medical Centre, Ulm, Germany
| | - Angela Silveira
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - P. Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan H. Smit
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Thomas H. Sparsø
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kathleen Stirrups
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ronald P. Stolk
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Heather M. Stringham
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Ann-Christine Syvänen
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sian-Tsung Tan
- Ealing Hospital NHS Trust, Middlesex, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Anke Tönjes
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | - Angelo Tremblay
- Department of Kinesiology, Laval University, Québec City, Québec, Canada
| | | | - Peter J. van der Most
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- School of Nutrition, Laval University, Québec City, Québec,Canada
| | - Judith M. Vonk
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, Munich, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Roman Wennauer
- Department of Clinical Chemistry, Ulm University Medical Centre, Ulm, Germany
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Tom Wilsgaard
- Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - M. Carola Zillikens
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Suzanne C. van Dijk
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja M. van Schoor
- EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
- VUMC, Department of Epidemiology and Biostatistics, Amsterdam, The Netherlands
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute Netherlands-Netherlands Heart Institute, Utrecht, The Netherlands
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Paul I. W. de Bakker
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Epidemiology, University Medical Center, Utrecht, The Netherlands
| | | | - John Beilby
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia,Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Carsten A. Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Bernhard O. Boehm
- Department of Internal Medicine I, Ulm University Medical Centre, Ulm, Germany
- Imperial College London, London, United Kingdom
- Lee Kong Chian School of Medicine, Singapore, Singapore
- Nanyang Technological University, Singapore, Singapore
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Stefan R. Bornstein
- Department of Medicine III, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Erwin P. Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Ealing Hospital NHS Trust, Middlesex, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Program in Biostatistics and Biomathematics, Divison of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts,United States of America
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato, Italy
- University of Sassari, Sassari, Italy
| | - Daniele Cusi
- Department of Health Sciences, University of Milan, Milan, Italy
- Institute of Biomedical Technologies, National Institute of Research, Segrate-Milano, Italy
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Jeanette Erdmann
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
- Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany
| | - Johan G. Eriksson
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Denis A. Evans
- Rush Institute for Healthy Aging and Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Farrall
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovacular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Lude Franke
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, Netherlands
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health & Clinical Medicine, Umeå University Hospital, Umeå, Sweden, Umeå, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Philippe Froguel
- CNRS UMR 8199, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille 2, Lille, France
| | - Ron T. Gansevoort
- University of Groningen, University Medical Center Groningen, Department of Medicine, Groningen, Netherlands
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ulf Gyllensten
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Hamsten
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute Netherlands-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Christian Hengstenberg
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, München, Germany
| | - Andrew A Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy, Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frank Hu
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Heikki V. Huikuri
- Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Kristian Hveem
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Joanne M. Jordan
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chaper Hill, North Carolina, United States of America
| | - Antti Jula
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Eero Kajantie
- Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sekar Kathiresan
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Lambertus A. L. M. Kiemeney
- Radboud university medical center, Radboud Institute for Health Sciences, Department for Health Evidence, Nijmegen, The Netherlands
- Radboud university medical center, Radboud Institute for Health Sciences, Department of Urology, Nijmegen, The Netherlands
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL, London, United Kingdom
| | - Paul B. Knekt
- National Institute for Health and Welfare, Helsinki, Finland
| | - Heikki A. Koistinen
- National Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Helsinki University Central Hospital, Department of Medicine and Abdominal Center: Endocrinology, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Seppo Koskinen
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Winfried Maerz
- Vth Department of Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nicholas G Martin
- QIMR Bergofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Physiology, Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere, Finland
| | - Guillaume Lettre
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Marja-Liisa Lokki
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Pekka Mäntyselkä
- Primary Health Care Unit, Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Primary Health Care Unit, Kuopio University Hospital, Kuopio, Finland
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Braxton D. Mitchell
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, United States of America
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, United States of America
| | - Frans L. Moll
- Department of Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeffrey C. Murray
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| | - Arthur W. Musk
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Markku S. Nieminen
- HUCH Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital, Helsinki, Finland
| | - Inger Njølstad
- Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Albertine J. Oldehinkel
- University of Groningen, University Medical Center, Interdisciplinary Center Psychopathology and Emotion Regulation, Groningen, The Netherlands
| | - Ben A. Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lyle J Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gerard Pasterkamp
- Experimental Cardiology and laboratory of clinical chemistry, UMCU, Utrecht, The Netherlands
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brenda W. Penninx
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Markus Perola
- Estonian Genome Center, Univeristy of Tartu, Tartu, Estonia
- National Institute for Health and Welfare, Department of Chronic Disease Prevention, Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Annette Peters
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, Munich, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Ozren Polašek
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy, Affiliated Institute of the University of Lübeck, Lübeck, Germany
- Department of Neurology, General Central Hospital, Bolzano, Italy
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Departments of Epidemiology and Health Services, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seatte, Washington, United States of America
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thomas Quertermous
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts,United States of America
| | - John D. Rioux
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Fernando Rivadeneira
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jerome I. Rotter
- Los Angeles BioMedical Resesarch Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Hester M. den Ruijter
- Experimental Cardiology and laboratory of clinical chemistry, UMCU, Utrecht, The Netherlands
| | - Juha Saltevo
- Department of Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, München, Germany
| | - Peter E. H. Schwarz
- Department of Medicine III, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Alan R. Shuldiner
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Dept of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Geriatric Research and Education Clinical Center, Vetrans Administration Medical Center, Baltimore, Maryland, United States of America
| | - Juha Sinisalo
- HUCH Heart and Lung Center, Division of Cardiology, Helsinki University Central Hospital, Helsinki, Finland
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Thorkild I. A. Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, Frederiksberg, Denmark
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Jan A. Staessen
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- R & D VitaK Group, Maastricht University, Maastricht, The Netherlands
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
| | - Jaakko Tuomilehto
- Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
- Centre for Vascular Prevention, Danube-University Krems, Krems, Austria
- Instituto de Investigacion Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - André G. Uitterlinden
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Matti Uusitupa
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Finland
- Research Unit, Kuopio University Hospital, Kuopio, Finland
| | - André L. M. Verbeek
- Radboud university medical center, Radboud Institute for Health Sciences, Department for Health Evidence, Nijmegen, The Netherlands
| | - Sita H. Vermeulen
- Radboud university medical center, Radboud Institute for Health Sciences, Department for Health Evidence, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Gérard Waeber
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Mark Walker
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Institute of Cellular Medicine, Newcastle University, Newcastle, United Kingdom
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Hugh Watkins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovacular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
| | - CHARGE Consortium
- The Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium
| | | | | | | | | | - MAGIC Consortium
- The Meta-Analyses of Glucose and Insulin-related traits Consortium
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Deborah J. Clegg
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - L. Adrienne Cupples
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Cashell E. Jaquish
- National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - D. C. Rao
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Goncalo R. Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Themistocles L. Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science Addenbrooke’s Hospital, Cambridge, United Kingdom
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Boehnke
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Panos Deloukas
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge, United Kingdom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Caroline S. Fox
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts,United States of America
| | - Leif C. Groop
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, Malmö, Sweden
- Finnish Institute for Molecular Medicine (FIMM), Helsinki University, Helsinki, Finland
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Erik Ingelsson
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Jeffrey R. O'Connell
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Dept of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, London, United Kingdom
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cecilia M. Lindgren
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University Regensburg, Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Kari E. North
- Carolina Center for Genome Sciences and Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Ruth J. F. Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
139
|
Cordell HJ, Han Y, Mells GF, Li Y, Hirschfield GM, Greene CS, Xie G, Juran BD, Zhu D, Qian DC, Floyd JAB, Morley KI, Prati D, Lleo A, Cusi D, Gershwin ME, Anderson CA, Lazaridis KN, Invernizzi P, Seldin MF, Sandford RN, Amos CI, Siminovitch KA. International genome-wide meta-analysis identifies new primary biliary cirrhosis risk loci and targetable pathogenic pathways. Nat Commun 2015; 6:8019. [PMID: 26394269 PMCID: PMC4580981 DOI: 10.1038/ncomms9019] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/03/2015] [Indexed: 12/16/2022] Open
Abstract
Primary biliary cirrhosis (PBC) is a classical autoimmune liver disease for which effective immunomodulatory therapy is lacking. Here we perform meta-analyses of discovery data sets from genome-wide association studies of European subjects (n=2,764 cases and 10,475 controls) followed by validation genotyping in an independent cohort (n=3,716 cases and 4,261 controls). We discover and validate six previously unknown risk loci for PBC (Pcombined<5 × 10(-8)) and used pathway analysis to identify JAK-STAT/IL12/IL27 signalling and cytokine-cytokine pathways, for which relevant therapies exist.
Collapse
Affiliation(s)
- Heather J. Cordell
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Younghun Han
- Center for Genomic Medicine, Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03755, USA
| | - George F. Mells
- Academic Department of Medical Genetics, Cambridge University, Cambridge CB2 0QQ, UK
| | - Yafang Li
- Center for Genomic Medicine, Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03755, USA
| | - Gideon M. Hirschfield
- Centre for Liver Research and NIHR Biomedical Research Unit, University of Birmingham, Birmingham B15 2TT, UK
| | - Casey S. Greene
- Department of Genetics, Institute for Quantitative Biomedical Sciences, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Gang Xie
- Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute and Toronto General Research Institute, Toronto, Ontario M5G 1X5, Canada
| | - Brian D. Juran
- Center for Basic Research in Digestive Diseases, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Dakai Zhu
- Center for Genomic Medicine, Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03755, USA
| | - David C. Qian
- Center for Genomic Medicine, Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03755, USA
| | - James A. B. Floyd
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Katherine I. Morley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
- Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London SE5 8AF, UK
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Ospedale Alessandro Manzoni, Lecco 23900, Italy
| | - Ana Lleo
- Center for Autoimmune Liver Diseases, IRCCS Instituto Clinico Humanitas, 20089 Rozzano, Italy
| | - Daniele Cusi
- Università degli Studi di Milano, Milan 20129, Italy
- Istituto di Tecnologie Biomediche Consiglio Nazionale delle Ricerche, Via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | | | - Carl A. Anderson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | | | - Pietro Invernizzi
- Center for Autoimmune Liver Diseases, IRCCS Instituto Clinico Humanitas, 20089 Rozzano, Italy
- University of California – Davis, Davis, California 95616, USA
| | | | - Richard N. Sandford
- Academic Department of Medical Genetics, Cambridge University, Cambridge CB2 0QQ, UK
| | - Christopher I. Amos
- Center for Genomic Medicine, Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03755, USA
| | - Katherine A. Siminovitch
- Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute and Toronto General Research Institute, Toronto, Ontario M5G 1X5, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada M5G 1X5
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada M5G 1X5
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5G 1X5
| |
Collapse
|
140
|
Sung YJ, Basson J, Cheng N, Nguyen KDH, Nandakumar P, Hunt SC, Arnett DK, Dávila-Román VG, Rao DC, Chakravarti A. The role of rare variants in systolic blood pressure: analysis of ExomeChip data in HyperGEN African Americans. Hum Hered 2015; 79:20-7. [PMID: 25765051 DOI: 10.1159/000375373] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/20/2015] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular diseases are among the most significant health problems in the United States today, with their major risk factor, hypertension, disproportionately affecting African Americans (AAs). Although GWAS have identified dozens of common variants associated with blood pressure (BP) and hypertension in European Americans, these variants collectively explain <2.5% of BP variance, and most of the genetic variants remain yet to be identified. Here, we report the results from rare-variant analysis of systolic BP using 94,595 rare and low-frequency variants (minor allele frequency, MAF, <5%) from the Illumina exome array genotyped in 2,045 HyperGEN AAs. In addition to single-variant analysis, 4 gene-level association tests were used for analysis: burden and family-based SKAT tests using MAF cutoffs of 1 and 5%. The gene-based methods often provided lower p values than the single-variant approach. Some consistency was observed across these 4 gene-based analysis options. While neither the gene-based analyses nor the single-variant analysis produced genome-wide significant results, the top signals, which had supporting evidence from multiple gene-based methods, were of borderline significance. Though additional molecular validations are required, 6 of the 16 most promising genes are biologically plausible with physiological connections to BP regulation.
Collapse
|
141
|
Reed E, Nunez S, Kulp D, Qian J, Reilly MP, Foulkes AS. A guide to genome-wide association analysis and post-analytic interrogation. Stat Med 2015; 34:3769-92. [PMID: 26343929 PMCID: PMC5019244 DOI: 10.1002/sim.6605] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/09/2015] [Accepted: 07/06/2015] [Indexed: 01/14/2023]
Abstract
This tutorial is a learning resource that outlines the basic process and provides specific software tools for implementing a complete genome‐wide association analysis. Approaches to post‐analytic visualization and interrogation of potentially novel findings are also presented. Applications are illustrated using the free and open‐source R statistical computing and graphics software environment, Bioconductor software for bioinformatics and the UCSC Genome Browser. Complete genome‐wide association data on 1401 individuals across 861,473 typed single nucleotide polymorphisms from the PennCATH study of coronary artery disease are used for illustration. All data and code, as well as additional instructional resources, are publicly available through the Open Resources in Statistical Genomics project: http://www.stat-gen.org. © 2015 The Authors. Statistics in Medicine Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Eric Reed
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| | - Sara Nunez
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| | - David Kulp
- Department of Computer Science, University of Massachusetts, Amherst, MA, U.S.A
| | - Jing Qian
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA, U.S.A
| | - Muredach P Reilly
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Andrea S Foulkes
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, U.S.A
| |
Collapse
|
142
|
Hayes MG, Urbanek M, Ehrmann DA, Armstrong LL, Lee JY, Sisk R, Karaderi T, Barber TM, McCarthy MI, Franks S, Lindgren CM, Welt CK, Diamanti-Kandarakis E, Panidis D, Goodarzi MO, Azziz R, Zhang Y, James RG, Olivier M, Kissebah AH, Stener-Victorin E, Legro RS, Dunaif A. Genome-wide association of polycystic ovary syndrome implicates alterations in gonadotropin secretion in European ancestry populations. Nat Commun 2015; 6:7502. [PMID: 26284813 PMCID: PMC4557132 DOI: 10.1038/ncomms8502] [Citation(s) in RCA: 251] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 05/14/2015] [Indexed: 12/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common, highly heritable complex disorder of unknown aetiology characterized by hyperandrogenism, chronic anovulation and defects in glucose homeostasis. Increased luteinizing hormone relative to follicle-stimulating hormone secretion, insulin resistance and developmental exposure to androgens are hypothesized to play a causal role in PCOS. Here we map common genetic susceptibility loci in European ancestry women for the National Institutes of Health PCOS phenotype, which confers the highest risk for metabolic morbidities, as well as reproductive hormone levels. Three loci reach genome-wide significance in the case–control meta-analysis, two novel loci mapping to chr 8p32.1 and chr 11p14.1, and a chr 9q22.32 locus previously found in Chinese PCOS. The same chr 11p14.1 SNP, rs11031006, in the region of the follicle-stimulating hormone B polypeptide (FSHB) gene strongly associates with PCOS diagnosis and luteinizing hormone levels. These findings implicate neuroendocrine changes in disease pathogenesis. Polycystic Ovary Sydrome is a highly heritable, complex reproductive disorder with unknown underlying genetic factors. Here Hayes and Urbanek et al. identify three loci in European women strongly associated with neuroendocrine changes and disease susceptibility.
Collapse
Affiliation(s)
- M Geoffrey Hayes
- 1] Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [3] Department of Anthropology, Northwestern University, Evanston, Illinois 60208, USA
| | - Margrit Urbanek
- 1] Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - David A Ehrmann
- Section of Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois 60637, USA
| | - Loren L Armstrong
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Ji Young Lee
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Ryan Sisk
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Tugce Karaderi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Thomas M Barber
- Warwick Medical School, University of Warwick, Warwick CV4 7AL, UK
| | - Mark I McCarthy
- 1] Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK [2] Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK [3] Oxford NIHR Biomedical Research Centre, Churchill Hospital, Headington OX3 7LE, UK
| | - Stephen Franks
- Institute of Reproductive &Developmental Biology, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Cecilia M Lindgren
- 1] Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK [2] Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, Utah 84112, USA
| | | | - Dimitrios Panidis
- Division of Endocrinology and Human Reproduction, 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki 54124, Greece
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Ricardo Azziz
- Departments of Obstetrics and Gynecology and Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Yi Zhang
- 1] TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA [2] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Roland G James
- 1] TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA [2] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Michael Olivier
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas 78256, USA
| | - Ahmed H Kissebah
- 1] TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA [2] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | - Richard S Legro
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Andrea Dunaif
- 1] Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
143
|
Hancock DB, Wang JC, Gaddis NC, Levy JL, Saccone NL, Stitzel JA, Goate A, Bierut LJ, Johnson EO. A multiancestry study identifies novel genetic associations with CHRNA5 methylation in human brain and risk of nicotine dependence. Hum Mol Genet 2015. [PMID: 26220977 DOI: 10.1093/hmg/ddv303] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Nicotine dependence is influenced by chromosome 15q25.1 single nucleotide polymorphisms (SNPs), including the missense SNP rs16969968 that alters function of the α5 nicotinic acetylcholine receptor (CHRNA5) and noncoding SNPs that regulate CHRNA5 mRNA expression. We tested for cis-methylation quantitative trait loci (cis-meQTLs) using SNP genotypes and DNA methylation levels measured across the IREB2-HYKK-PSMA4-CHRNA5-CHRNA3-CHRNB4 genes on chromosome 15q25.1 in the BrainCloud and Brain QTL cohorts [total N = 175 European-Americans and 65 African-Americans (AAs)]. We identified eight SNPs that were significantly associated with CHRNA5 methylation in prefrontal cortex: P ranging from 6.0 × 10(-10) to 5.6 × 10(-5). These SNP-methylation associations were also significant in frontal cortex, temporal cortex and pons: P ranging from 4.8 × 10(-12) to 3.4 × 10(-3). Of the eight cis-meQTL SNPs, only the intronic CHRNB4 SNP rs11636753 was associated with CHRNA5 methylation independently of the known SNP effects in prefrontal cortex, and it was the most significantly associated SNP with nicotine dependence across five independent cohorts (total N = 7858 European ancestry and 3238 AA participants): P = 6.7 × 10(-4), odds ratio (OR) [95% confidence interval (CI)] = 1.11 (1.05-1.18). The rs11636753 major allele (G) was associated with lower CHRNA5 DNA methylation, lower CHRNA5 mRNA expression and increased nicotine dependence risk. Haplotype analyses showed that rs11636753-G and the functional rs16969968-A alleles together increased risk of nicotine dependence more than each variant alone: P = 3.1 × 10(-12), OR (95% CI) = 1.32 (1.22-1.43). Our findings identify a novel regulatory SNP association with nicotine dependence and connect, for the first time, previously observed differences in CHRNA5 mRNA expression and nicotine dependence risk to underlying DNA methylation differences.
Collapse
Affiliation(s)
- Dana B Hancock
- Behavioral and Urban Health Program, Behavioral Health and Criminal Justice Division,
| | - Jen-Chyong Wang
- Department of Neuroscience and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | | | - Jerry A Stitzel
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309, USA
| | - Alison Goate
- Department of Neuroscience and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA and
| | - Eric O Johnson
- Fellow Program and Behavioral Health and Criminal Justice Division, Research Triangle Institute (RTI) International, Research Triangle Park, NC 27709, USA
| |
Collapse
|
144
|
Shin J, Bourdon C, Bernard M, Wilson MD, Reischl E, Waldenberger M, Ruggeri B, Schumann G, Desrivieres S, Leemans A, Abrahamowicz M, Leonard G, Richer L, Bouchard L, Gaudet D, Paus T, Pausova Z. Layered genetic control of DNA methylation and gene expression: a locus of multiple sclerosis in healthy individuals. Hum Mol Genet 2015. [PMID: 26220975 DOI: 10.1093/hmg/ddv294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DNA methylation may contribute to the etiology of complex genetic disorders through its impact on genome integrity and gene expression; it is modulated by DNA-sequence variants, named methylation quantitative trait loci (meQTLs). Most meQTLs influence methylation of a few CpG dinucleotides within short genomic regions (<3 kb). Here, we identified a layered genetic control of DNA methylation at numerous CpGs across a long 300 kb genomic region. This control involved a single long-range meQTL and multiple local meQTLs. The long-range meQTL explained up to 75% of variance in methylation of CpGs located over extended areas of the 300 kb region. The meQTL was identified in four samples (P = 2.8 × 10(-17), 3.1 × 10(-31), 4.0 × 10(-71) and 5.2 × 10(-199)), comprising a total of 2796 individuals. The long-range meQTL was strongly associated not only with DNA methylation but also with mRNA expression of several genes within the 300 kb region (P = 7.1 × 10(-18)-1.0 × 10(-123)). The associations of the meQTL with gene expression became attenuated when adjusted for DNA methylation (causal inference test: P = 2.4 × 10(-13)-7.1 × 10(-20)), indicating coordinated regulation of DNA methylation and gene expression. Further, the long-range meQTL was found to be in linkage disequilibrium with the most replicated locus of multiple sclerosis, a disease affecting primarily the brain white matter. In middle-aged adults free of the disease, we observed that the risk allele was associated with subtle structural properties of the brain white matter found in multiple sclerosis (P = 0.02). In summary, we identified a long-range meQTL that controls methylation and expression of several genes and may be involved in increasing brain vulnerability to multiple sclerosis.
Collapse
Affiliation(s)
- Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Celine Bourdon
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Manon Bernard
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Michael D Wilson
- The Hospital for Sick Children, University of Toronto, Toronto, Canada, Department of Molecular Genetics
| | - Eva Reischl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Munich, Germany
| | - Barbara Ruggeri
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gunter Schumann
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sylvane Desrivieres
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexander Leemans
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | - Gabriel Leonard
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Louis Richer
- Department of Psychology, Université du Québec à Chicoutimi, Chicoutimi, Canada
| | - Luigi Bouchard
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, Canada, ECOGENE-21 and Lipid Clinic, Chicoutimi Hospital, Chicoutimi, Canada
| | - Daniel Gaudet
- ECOGENE-21 and Lipid Clinic, Chicoutimi Hospital, Chicoutimi, Canada, Department of Medicine, Université de Montréal, Montréal, Canada and
| | - Tomas Paus
- Rotman Research Institute, University of Toronto, Toronto, Canada, Child Mind Institute, New York, NY, USA
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, Canada, Department of Physiology, Department of Nutritional Sciences,
| |
Collapse
|
145
|
Soave D, Corvol H, Panjwani N, Gong J, Li W, Boëlle PY, Durie PR, Paterson AD, Rommens JM, Strug LJ, Sun L. A Joint Location-Scale Test Improves Power to Detect Associated SNPs, Gene Sets, and Pathways. Am J Hum Genet 2015; 97:125-38. [PMID: 26140448 PMCID: PMC4572492 DOI: 10.1016/j.ajhg.2015.05.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/26/2015] [Indexed: 11/28/2022] Open
Abstract
Gene-based, pathway, and other multivariate association methods are motivated by the possibility of GxG and GxE interactions; however, accounting for such interactions is limited by the challenges associated with adequate modeling information. Here we propose an easy-to-implement joint location-scale (JLS) association testing framework for single-variant and multivariate analysis that accounts for interactions without explicitly modeling them. We apply the JLS method to a gene-set analysis of cystic fibrosis (CF) lung disease, which is influenced by multiple environmental and genetic factors. We identify and replicate an association between the constituents of the apical plasma membrane and CF lung disease (p = 0.0099 and p = 0.0180, respectively) and highlight a role for the SLC9A3-SLC9A3R1/2-EZR complex in contributing to CF lung disease. Many association studies could benefit from re-analysis with the JLS method that leverages complex genetic architecture for SNP, gene, and pathway identification. Analytical verification, simulation, and additional proof-of-principle applications support our approach.
Collapse
Affiliation(s)
- David Soave
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Harriet Corvol
- Assistance Publique-Hôpitaux de Paris (AP-HP), Trousseau Hospital, Pediatric Pulmonology Department; Institut National de la Santé et la Recherche Médicale (INSERM), UMR_S 938, CDR Saint-Antoine, 75012 Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, 75005 Paris, France
| | - Naim Panjwani
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jiafen Gong
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Weili Li
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Pierre-Yves Boëlle
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, 75005 Paris, France; AP-HP, Saint-Antoine Hospital, Biostatistics Department, INSERM, UMR_S 1136, 75012 Paris, France
| | - Peter R Durie
- Program in Physiology and Experimental Medicine, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Pediatrics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Andrew D Paterson
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Johanna M Rommens
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lisa J Strug
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
| | - Lei Sun
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; Department of Statistical Sciences, University of Toronto, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
146
|
Genetic variants in the ADAMTS13 and SUPT3H genes are associated with ADAMTS13 activity. Blood 2015; 125:3949-55. [DOI: 10.1182/blood-2015-02-629865] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/27/2015] [Indexed: 12/21/2022] Open
Abstract
Key Points
We identify rs41314453 as the strongest genetic predictor of ADAMTS13 activity, associated with a decrease of >20%. We present evidence of further independent associations with a common variant in SUPT3H, as well as 5 variants at the ADAMTS13 locus.
Collapse
|
147
|
Genome-Wide Meta-Analysis of Longitudinal Alcohol Consumption Across Youth and Early Adulthood. Twin Res Hum Genet 2015; 18:335-47. [PMID: 26081443 DOI: 10.1017/thg.2015.36] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The public health burden of alcohol is unevenly distributed across the life course, with levels of use, abuse, and dependence increasing across adolescence and peaking in early adulthood. Here, we leverage this temporal patterning to search for common genetic variants predicting developmental trajectories of alcohol consumption. Comparable psychiatric evaluations measuring alcohol consumption were collected in three longitudinal community samples (N=2,126, obs=12,166). Consumption-repeated measurements spanning adolescence and early adulthood were analyzed using linear mixed models, estimating individual consumption trajectories, which were then tested for association with Illumina 660W-Quad genotype data (866,099 SNPs after imputation and QC). Association results were combined across samples using standard meta-analysis methods. Four meta-analysis associations satisfied our pre-determined genome-wide significance criterion (FDR<0.1) and six others met our 'suggestive' criterion (FDR<0.2). Genome-wide significant associations were highly biological plausible, including associations within GABA transporter 1, SLC6A1 (solute carrier family 6, member 1), and exonic hits in LOC100129340 (mitofusin-1-like). Pathway analyses elaborated single marker results, indicating significant enriched associations to intuitive biological mechanisms, including neurotransmission, xenobiotic pharmacodynamics, and nuclear hormone receptors (NHR). These findings underscore the value of combining longitudinal behavioral data and genome-wide genotype information in order to study developmental patterns and improve statistical power in genomic studies.
Collapse
|
148
|
Cuellar-Partida G, Springelkamp H, Lucas SEM, Yazar S, Hewitt AW, Iglesias AI, Montgomery GW, Martin NG, Pennell CE, van Leeuwen EM, Verhoeven VJM, Hofman A, Uitterlinden AG, Ramdas WD, Wolfs RCW, Vingerling JR, Brown MA, Mills RA, Craig JE, Klaver CCW, van Duijn CM, Burdon KP, MacGregor S, Mackey DA. WNT10A exonic variant increases the risk of keratoconus by decreasing corneal thickness. Hum Mol Genet 2015; 24:5060-8. [PMID: 26049155 DOI: 10.1093/hmg/ddv211] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/02/2015] [Indexed: 11/13/2022] Open
Abstract
Keratoconus is a degenerative eye condition which results from thinning of the cornea and causes vision distortion. Treatments such as ultraviolet (UV) cross-linking have proved effective for management of keratoconus when performed in early stages of the disease. The central corneal thickness (CCT) is a highly heritable endophenotype of keratoconus, and it is estimated that up to 95% of its phenotypic variance is due to genetics. Genome-wide association efforts of CCT have identified common variants (i.e. minor allele frequency (MAF) >5%). However, these studies typically ignore the large set of exonic variants whose MAF is usually low. In this study, we performed a CCT exome-wide association analysis in a sample of 1029 individuals from a population-based study in Western Australia. We identified a genome-wide significant exonic variant rs121908120 (P = 6.63 × 10(-10)) in WNT10A. This gene is 437 kb from a gene previously associated with CCT (USP37). We showed in a conditional analysis that the WNT10A variant completely accounts for the signal previously seen at USP37. We replicated our finding in independent samples from the Brisbane Adolescent Twin Study, Twin Eye Study in Tasmania and the Rotterdam Study. Further, we genotyped rs121908120 in 621 keratoconus cases and compared the frequency to a sample of 1680 unscreened controls from the Queensland Twin Registry. We found that rs121908120 increases the risk of keratoconus two times (odds ratio 2.03, P = 5.41 × 10(-5)).
Collapse
Affiliation(s)
| | | | | | - Seyhan Yazar
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia
| | - Alex W Hewitt
- School of Medicine, Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia, Centre for Eye Research Australia, Melbourne University, Melbourne, Australia
| | | | | | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia and
| | - Craig E Pennell
- School of Women's and Infants' Health, University of Western Australia, Perth, Australia
| | | | | | - Albert Hofman
- Department of Epidemiology, Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, the Hague 2593 CE, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands, Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, the Hague 2593 CE, The Netherlands
| | | | | | | | - Matthew A Brown
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Australia
| | - Richard A Mills
- Department of Ophthalmology, Flinders University, Adelaide, SA, Australia
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, SA, Australia
| | | | | | | | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia,
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia
| |
Collapse
|
149
|
Richter HE, Whitehead N, Arya L, Ridgeway B, Allen-Brady K, Norton P, Sung V, Shepherd JP, Komesu Y, Gaddis N, Fraser MO, Tan-Kim J, Meikle S, Page GP. Genetic contributions to urgency urinary incontinence in women. J Urol 2015; 193:2020-7. [PMID: 25524241 PMCID: PMC4439377 DOI: 10.1016/j.juro.2014.12.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2014] [Indexed: 01/20/2023]
Abstract
PURPOSE We identify genetic variants associated with urgency urinary incontinence in postmenopausal women. MATERIALS AND METHODS A 2-stage genome-wide association analysis was conducted to identify variants associated with urgency urinary incontinence. The WHI GARNET substudy with 4,894 genotyped post-reproductive white women was randomly split into independent discovery and replication cohorts. Genome-wide imputation was performed using IMPUTE2 with the 1000 Genomes ALL Phase I integrated variant set as a reference. Controls reported no urgency urinary incontinence at enrollment or followup. Cases reported monthly or greater urgency urinary incontinence and leaked sufficiently to wet/soak underpants/clothes. Logistic regression models were used to predict urgency urinary incontinence case vs control status based on genotype, assuming additive inheritance. Age, obesity, diabetes and depression were included in the models as covariates. RESULTS Following quality control, 975,508 single nucleotide polymorphisms in 2,241 cases (discovery 1,102; replication 1,133) and 776 controls (discovery 405, replication 371) remained. Genotype imputation resulted in 9,077,347 single nucleotide polymorphisms and insertions/deletions with minor allele frequency greater than 0.01 available for analysis. Meta-analysis of the discovery and replication samples identified 6 loci on chromosomes 5, 10, 11, 12 and 18 associated with urgency urinary incontinence at p <10(-6). Of the loci 3 were within genes, the zinc finger protein 521 (ZFP521) gene on chromosome 18q11, the ADAMTS16 gene on chromosome 5p15 and the CIT gene on chromosome 12q24. The other 3 loci were intergenic. CONCLUSIONS Although environmental factors also likely contribute, this first exploratory genome-wide association study for urgency urinary incontinence suggests that genetic variants in the ZFP521, CIT and ADAMTS16 genes might account for some of the observed heritability of the condition.
Collapse
Affiliation(s)
| | - Nedra Whitehead
- Research Triangle International, Research Triangle Park, North Carolina
| | - Lily Arya
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - Vivian Sung
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | - Yuko Komesu
- University of New Mexico, Albuquerque, New Mexico
| | - Nathan Gaddis
- Research Triangle International, Research Triangle Park, North Carolina
| | | | | | - Susan Meikle
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| | - Grier P Page
- Research Triangle International, Research Triangle Park, North Carolina
| |
Collapse
|
150
|
Johansson Å, Eriksson N, Becker RC, Storey RF, Himmelmann A, Hagström E, Varenhorst C, Axelsson T, Barratt BJ, James SK, Katus HA, Steg PG, Syvänen AC, Wallentin L, Siegbahn A. NLRC4 Inflammasome Is an Important Regulator of Interleukin-18 Levels in Patients With Acute Coronary Syndromes. ACTA ACUST UNITED AC 2015; 8:498-506. [DOI: 10.1161/circgenetics.114.000724] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 02/16/2015] [Indexed: 01/10/2023]
Abstract
Background—
Interleukin 18 (IL-18) promotes atherosclerotic plaque formation and is increased in patients with acute coronary syndromes. However the relative contribution of genetic variants to the IL-18 levels has not been fully determined.
Methods and Results—
Baseline plasma IL-18 levels were measured in 16 633 patients with acute coronary syndrome, of whom 9340 had genetic data that passed genotype quality control. A 2-stage genome-wide association study was performed, followed by combined analyses using >10 million genotyped or imputed genetic markers. Single nucleotide polymorphisms at 3 loci (
IL18, NLRC4
, and
MROH6
) were identified (
P
<3.15×10
−8
) in the discovery cohort (n=3777) and replicated in the remaining patients (n=5563). In the pooled data (discovery+replication cohort), 7 independent associations, in 5 chromosomal regions, were associated with IL-18 levels (minimum
P
=6.99×10
–72
). Six single nucleotide polymorphisms are located in predicted promoter regions of which one disrupts a transcription factor binding site. One single nucleotide polymorphism in
NLRC4
is a rare missense variant, predicted to be deleterious to the protein. Altogether, the identified genetic variants explained 8% of the total variation in IL-18 levels in the cohort.
Conclusions—
Our results show that genetic variants play an important role in determining IL-18 levels in patients with acute coronary syndrome and we have identified genetic variants located in the IL-18 gene (
IL18
) or close to genes that are involved in procaspase-1 activation (
NLRC4
and
CARD16
,
CARD17
, and
CARD18
). These associations also highlight the importance of the NLRC4 inflammasome for IL-18 production in acute coronary syndrome patients.
Collapse
|