Your browser doesn't support javascript.
loading
Integration of multiethnic fine-mapping and genomic annotation to prioritize candidate functional SNPs at prostate cancer susceptibility regions.
Han, Ying; Hazelett, Dennis J; Wiklund, Fredrik; Schumacher, Fredrick R; Stram, Daniel O; Berndt, Sonja I; Wang, Zhaoming; Rand, Kristin A; Hoover, Robert N; Machiela, Mitchell J; Yeager, Merideth; Burdette, Laurie; Chung, Charles C; Hutchinson, Amy; Yu, Kai; Xu, Jianfeng; Travis, Ruth C; Key, Timothy J; Siddiq, Afshan; Canzian, Federico; Takahashi, Atsushi; Kubo, Michiaki; Stanford, Janet L; Kolb, Suzanne; Gapstur, Susan M; Diver, W Ryan; Stevens, Victoria L; Strom, Sara S; Pettaway, Curtis A; Al Olama, Ali Amin; Kote-Jarai, Zsofia; Eeles, Rosalind A; Yeboah, Edward D; Tettey, Yao; Biritwum, Richard B; Adjei, Andrew A; Tay, Evelyn; Truelove, Ann; Niwa, Shelley; Chokkalingam, Anand P; Isaacs, William B; Chen, Constance; Lindstrom, Sara; Le Marchand, Loic; Giovannucci, Edward L; Pomerantz, Mark; Long, Henry; Li, Fugen; Ma, Jing; Stampfer, Meir.
Afiliación
  • Han Y; Department of Preventive Medicine, Keck School of Medicine.
  • Hazelett DJ; Department of Preventive Medicine, Keck School of Medicine.
  • Wiklund F; Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.
  • Schumacher FR; Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center.
  • Stram DO; Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center.
  • Berndt SI; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Wang Z; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA.
  • Rand KA; Department of Preventive Medicine, Keck School of Medicine.
  • Hoover RN; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Machiela MJ; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Yeager M; Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA.
  • Burdette L; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA.
  • Chung CC; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Hutchinson A; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA.
  • Yu K; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
  • Xu J; Program for Personalized Cancer Care and Department of Surgery, NorthShore University HealthSystem, Evanston, IL, USA.
  • Travis RC; Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
  • Key TJ; Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
  • Siddiq A; Department of Genomics of Common Disease, School of Public Health.
  • Canzian F; Genomic Epidemiology Group, German Cancer Research Center, Heidelberg, Germany.
  • Takahashi A; Laboratory for Statistical Analysis.
  • Kubo M; Laboratory for Genotyping Development.
  • Stanford JL; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA.
  • Kolb S; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
  • Gapstur SM; Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA.
  • Diver WR; Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA.
  • Stevens VL; Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA.
  • Strom SS; Department of Epidemiology.
  • Pettaway CA; Department of Urology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
  • Al Olama AA; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
  • Kote-Jarai Z; The Institute of Cancer Research, London, UK.
  • Eeles RA; The Institute of Cancer Research, London, UK, Royal Marsden National Health Services (NHS) Foundation Trust, London and Sutton, UK.
  • Yeboah ED; Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana.
  • Tettey Y; Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana.
  • Biritwum RB; Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana.
  • Adjei AA; Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana.
  • Tay E; Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana.
  • Truelove A; Westat, Rockville, MD, USA.
  • Niwa S; Westat, Rockville, MD, USA.
  • Chokkalingam AP; School of Public Health, University of California, Berkeley, CA, USA.
  • Isaacs WB; James Buchanan Brady Urological Institute, Johns Hopkins Hospital and Medical Institution, Baltimore, MD, USA.
  • Chen C; Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology.
  • Lindstrom S; Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology.
  • Le Marchand L; Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA.
  • Giovannucci EL; Department of Nutrition, Department of Epidemiology.
  • Pomerantz M; Department of Medical Oncology.
  • Long H; Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
  • Li F; Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
  • Ma J; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
  • Stampfer M; Department of Nutrition, Department of Epidemiology.
Hum Mol Genet ; 24(19): 5603-18, 2015 Oct 01.
Article en En | MEDLINE | ID: mdl-26162851
ABSTRACT
Interpretation of biological mechanisms underlying genetic risk associations for prostate cancer is complicated by the relatively large number of risk variants (n = 100) and the thousands of surrogate SNPs in linkage disequilibrium. Here, we combined three distinct approaches multiethnic fine-mapping, putative functional annotation (based upon epigenetic data and genome-encoded features), and expression quantitative trait loci (eQTL) analyses, in an attempt to reduce this complexity. We examined 67 risk regions using genotyping and imputation-based fine-mapping in populations of European (cases/controls 8600/6946), African (cases/controls 5327/5136), Japanese (cases/controls 2563/4391) and Latino (cases/controls 1034/1046) ancestry. Markers at 55 regions passed a region-specific significance threshold (P-value cutoff range 3.9 × 10(-4)-5.6 × 10(-3)) and in 30 regions we identified markers that were more significantly associated with risk than the previously reported variants in the multiethnic sample. Novel secondary signals (P < 5.0 × 10(-6)) were also detected in two regions (rs13062436/3q21 and rs17181170/3p12). Among 666 variants in the 55 regions with P-values within one order of magnitude of the most-associated marker, 193 variants (29%) in 48 regions overlapped with epigenetic or other putative functional marks. In 11 of the 55 regions, cis-eQTLs were detected with nearby genes. For 12 of the 55 regions (22%), the most significant region-specific, prostate-cancer associated variant represented the strongest candidate functional variant based on our annotations; the number of regions increased to 20 (36%) and 27 (49%) when examining the 2 and 3 most significantly associated variants in each region, respectively. These results have prioritized subsets of candidate variants for downstream functional evaluation.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Próstata / Hispánicos o Latinos / Polimorfismo de Nucleótido Simple / Población Negra / Pueblo Asiatico / Población Blanca Tipo de estudio: Prognostic_studies Límite: Humans / Male Idioma: En Revista: Hum Mol Genet Asunto de la revista: BIOLOGIA MOLECULAR / GENETICA MEDICA Año: 2015 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Próstata / Hispánicos o Latinos / Polimorfismo de Nucleótido Simple / Población Negra / Pueblo Asiatico / Población Blanca Tipo de estudio: Prognostic_studies Límite: Humans / Male Idioma: En Revista: Hum Mol Genet Asunto de la revista: BIOLOGIA MOLECULAR / GENETICA MEDICA Año: 2015 Tipo del documento: Article