132 results on '"Kimberly F Doheny"'
Search Results
2. Novel genetic locus implicated for HIV-1 acquisition with putative regulatory links to HIV replication and infectivity: a genome-wide association study.
- Author
-
Eric O Johnson, Dana B Hancock, Nathan C Gaddis, Joshua L Levy, Grier Page, Scott P Novak, Cristie Glasheen, Nancy L Saccone, John P Rice, Michael P Moreau, Kimberly F Doheny, Jane M Romm, Andrew I Brooks, Bradley E Aouizerat, Laura J Bierut, and Alex H Kral
- Subjects
Medicine ,Science - Abstract
Fifty percent of variability in HIV-1 susceptibility is attributable to host genetics. Thus identifying genetic associations is essential to understanding pathogenesis of HIV-1 and important for targeting drug development. To date, however, CCR5 remains the only gene conclusively associated with HIV acquisition. To identify novel host genetic determinants of HIV-1 acquisition, we conducted a genome-wide association study among a high-risk sample of 3,136 injection drug users (IDUs) from the Urban Health Study (UHS). In addition to being IDUs, HIV-controls were frequency-matched to cases on environmental exposures to enhance detection of genetic effects. We tested independent replication in the Women's Interagency HIV Study (N=2,533). We also examined publicly available gene expression data to link SNPs associated with HIV acquisition to known mechanisms affecting HIV replication/infectivity. Analysis of the UHS nominated eight genetic regions for replication testing. SNP rs4878712 in FRMPD1 met multiple testing correction for independent replication (P=1.38x10(-4)), although the UHS-WIHS meta-analysis p-value did not reach genome-wide significance (P=4.47x10(-7) vs. P
- Published
- 2015
- Full Text
- View/download PDF
3. Genome-wide meta-analysis of homocysteine and methionine metabolism identifies five one carbon metabolism loci and a novel association of ALDH1L1 with ischemic stroke.
- Author
-
Stephen R Williams, Qiong Yang, Fang Chen, Xuan Liu, Keith L Keene, Paul Jacques, Wei-Min Chen, Galit Weinstein, Fang-Chi Hsu, Alexa Beiser, Liewei Wang, Ebony Bookman, Kimberly F Doheny, Philip A Wolf, Michelle Zilka, Jacob Selhub, Sarah Nelson, Stephanie M Gogarten, Bradford B Worrall, Sudha Seshadri, Michèle M Sale, Genomics and Randomized Trials Network, and Framingham Heart Study
- Subjects
Genetics ,QH426-470 - Abstract
Circulating homocysteine levels (tHcy), a product of the folate one carbon metabolism pathway (FOCM) through the demethylation of methionine, are heritable and are associated with an increased risk of common diseases such as stroke, cardiovascular disease (CVD), cancer and dementia. The FOCM is the sole source of de novo methyl group synthesis, impacting many biological and epigenetic pathways. However, the genetic determinants of elevated tHcy (hyperhomocysteinemia), dysregulation of methionine metabolism and the underlying biological processes remain unclear. We conducted independent genome-wide association studies and a meta-analysis of methionine metabolism, characterized by post-methionine load test tHcy, in 2,710 participants from the Framingham Heart Study (FHS) and 2,100 participants from the Vitamin Intervention for Stroke Prevention (VISP) clinical trial, and then examined the association of the identified loci with incident stroke in FHS. Five genes in the FOCM pathway (GNMT [p = 1.60 × 10(-63)], CBS [p = 3.15 × 10(-26)], CPS1 [p = 9.10 × 10(-13)], ALDH1L1 [p = 7.3 × 10(-13)] and PSPH [p = 1.17 × 10(-16)]) were strongly associated with the difference between pre- and post-methionine load test tHcy levels (ΔPOST). Of these, one variant in the ALDH1L1 locus, rs2364368, was associated with incident ischemic stroke. Promoter analyses reveal genetic and epigenetic differences that may explain a direct effect on GNMT transcription and a downstream affect on methionine metabolism. Additionally, a genetic-score consisting of the five significant loci explains 13% of the variance of ΔPOST in FHS and 6% of the variance in VISP. Association between variants in FOCM genes with ΔPOST suggest novel mechanisms that lead to differences in methionine metabolism, and possibly the epigenome, impacting disease risk. These data emphasize the importance of a concerted effort to understand regulators of one carbon metabolism as potential therapeutic targets.
- Published
- 2014
- Full Text
- View/download PDF
4. Fine-mapping analysis including over 254,000 East Asian and European descendants identifies 136 putative colorectal cancer susceptibility genes
- Author
-
Zhishan Chen, Xingyi Guo, Ran Tao, Jeroen R. Huyghe, Philip J. Law, Ceres Fernandez-Rozadilla, Jie Ping, Guochong Jia, Jirong Long, Chao Li, Quanhu Shen, Yuhan Xie, Maria N. Timofeeva, Minta Thomas, Stephanie L. Schmit, Virginia Díez-Obrero, Matthew Devall, Ferran Moratalla-Navarro, Juan Fernandez-Tajes, Claire Palles, Kitty Sherwood, Sarah E. W. Briggs, Victoria Svinti, Kevin Donnelly, Susan M. Farrington, James Blackmur, Peter G. Vaughan-Shaw, Xiao-Ou Shu, Yingchang Lu, Peter Broderick, James Studd, Tabitha A. Harrison, David V. Conti, Fredrick R. Schumacher, Marilena Melas, Gad Rennert, Mireia Obón-Santacana, Vicente Martín-Sánchez, Jae Hwan Oh, Jeongseon Kim, Sun Ha Jee, Keum Ji Jung, Sun-Seog Kweon, Min-Ho Shin, Aesun Shin, Yoon-Ok Ahn, Dong-Hyun Kim, Isao Oze, Wanqing Wen, Keitaro Matsuo, Koichi Matsuda, Chizu Tanikawa, Zefang Ren, Yu-Tang Gao, Wei-Hua Jia, John L. Hopper, Mark A. Jenkins, Aung Ko Win, Rish K. Pai, Jane C. Figueiredo, Robert W. Haile, Steven Gallinger, Michael O. Woods, Polly A. Newcomb, David Duggan, Jeremy P. Cheadle, Richard Kaplan, Rachel Kerr, David Kerr, Iva Kirac, Jan Böhm, Jukka-Pekka Mecklin, Pekka Jousilahti, Paul Knekt, Lauri A. Aaltonen, Harri Rissanen, Eero Pukkala, Johan G. Eriksson, Tatiana Cajuso, Ulrika Hänninen, Johanna Kondelin, Kimmo Palin, Tomas Tanskanen, Laura Renkonen-Sinisalo, Satu Männistö, Demetrius Albanes, Stephanie J. Weinstein, Edward Ruiz-Narvaez, Julie R. Palmer, Daniel D. Buchanan, Elizabeth A. Platz, Kala Visvanathan, Cornelia M. Ulrich, Erin Siegel, Stefanie Brezina, Andrea Gsur, Peter T. Campbell, Jenny Chang-Claude, Michael Hoffmeister, Hermann Brenner, Martha L. Slattery, John D. Potter, Kostas K. Tsilidis, Matthias B. Schulze, Marc J. Gunter, Neil Murphy, Antoni Castells, Sergi Castellví-Bel, Leticia Moreira, Volker Arndt, Anna Shcherbina, D. Timothy Bishop, Graham G. Giles, Melissa C. Southey, Gregory E. Idos, Kevin J. McDonnell, Zomoroda Abu-Ful, Joel K. Greenson, Katerina Shulman, Flavio Lejbkowicz, Kenneth Offit, Yu-Ru Su, Robert Steinfelder, Temitope O. Keku, Bethany van Guelpen, Thomas J. Hudson, Heather Hampel, Rachel Pearlman, Sonja I. Berndt, Richard B. Hayes, Marie Elena Martinez, Sushma S. Thomas, Paul D. P. Pharoah, Susanna C. Larsson, Yun Yen, Heinz-Josef Lenz, Emily White, Li Li, Kimberly F. Doheny, Elizabeth Pugh, Tameka Shelford, Andrew T. Chan, Marcia Cruz-Correa, Annika Lindblom, David J. Hunter, Amit D. Joshi, Clemens Schafmayer, Peter C. Scacheri, Anshul Kundaje, Robert E. Schoen, Jochen Hampe, Zsofia K. Stadler, Pavel Vodicka, Ludmila Vodickova, Veronika Vymetalkova, Christopher K. Edlund, W. James Gauderman, David Shibata, Amanda Toland, Sanford Markowitz, Andre Kim, Stephen J. Chanock, Franzel van Duijnhoven, Edith J. M. Feskens, Lori C. Sakoda, Manuela Gago-Dominguez, Alicja Wolk, Barbara Pardini, Liesel M. FitzGerald, Soo Chin Lee, Shuji Ogino, Stephanie A. Bien, Charles Kooperberg, Christopher I. Li, Yi Lin, Ross Prentice, Conghui Qu, Stéphane Bézieau, Taiki Yamaji, Norie Sawada, Motoki Iwasaki, Loic Le Marchand, Anna H. Wu, Chenxu Qu, Caroline E. McNeil, Gerhard Coetzee, Caroline Hayward, Ian J. Deary, Sarah E. Harris, Evropi Theodoratou, Stuart Reid, Marion Walker, Li Yin Ooi, Ken S. Lau, Hongyu Zhao, Li Hsu, Qiuyin Cai, Malcolm G. Dunlop, Stephen B. Gruber, Richard S. Houlston, Victor Moreno, Graham Casey, Ulrike Peters, Ian Tomlinson, and Wei Zheng
- Subjects
Science - Abstract
Abstract Genome-wide association studies (GWAS) have identified more than 200 common genetic variants independently associated with colorectal cancer (CRC) risk, but the causal variants and target genes are mostly unknown. We sought to fine-map all known CRC risk loci using GWAS data from 100,204 cases and 154,587 controls of East Asian and European ancestry. Our stepwise conditional analyses revealed 238 independent association signals of CRC risk, each with a set of credible causal variants (CCVs), of which 28 signals had a single CCV. Our cis-eQTL/mQTL and colocalization analyses using colorectal tissue-specific transcriptome and methylome data separately from 1299 and 321 individuals, along with functional genomic investigation, uncovered 136 putative CRC susceptibility genes, including 56 genes not previously reported. Analyses of single-cell RNA-seq data from colorectal tissues revealed 17 putative CRC susceptibility genes with distinct expression patterns in specific cell types. Analyses of whole exome sequencing data provided additional support for several target genes identified in this study as CRC susceptibility genes. Enrichment analyses of the 136 genes uncover pathways not previously linked to CRC risk. Our study substantially expanded association signals for CRC and provided additional insight into the biological mechanisms underlying CRC development.
- Published
- 2024
- Full Text
- View/download PDF
5. Common variants at 9p21 and 8q22 are associated with increased susceptibility to optic nerve degeneration in glaucoma.
- Author
-
Janey L Wiggs, Brian L Yaspan, Michael A Hauser, Jae H Kang, R Rand Allingham, Lana M Olson, Wael Abdrabou, Bao J Fan, Dan Y Wang, Wendy Brodeur, Donald L Budenz, Joseph Caprioli, Andrew Crenshaw, Kristy Crooks, Elizabeth Delbono, Kimberly F Doheny, David S Friedman, Douglas Gaasterland, Terry Gaasterland, Cathy Laurie, Richard K Lee, Paul R Lichter, Stephanie Loomis, Yutao Liu, Felipe A Medeiros, Cathy McCarty, Daniel Mirel, Sayoko E Moroi, David C Musch, Anthony Realini, Frank W Rozsa, Joel S Schuman, Kathleen Scott, Kuldev Singh, Joshua D Stein, Edward H Trager, Paul Vanveldhuisen, Douglas Vollrath, Gadi Wollstein, Sachiko Yoneyama, Kang Zhang, Robert N Weinreb, Jason Ernst, Manolis Kellis, Tomohiro Masuda, Don Zack, Julia E Richards, Margaret Pericak-Vance, Louis R Pasquale, and Jonathan L Haines
- Subjects
Genetics ,QH426-470 - Abstract
Optic nerve degeneration caused by glaucoma is a leading cause of blindness worldwide. Patients affected by the normal-pressure form of glaucoma are more likely to harbor risk alleles for glaucoma-related optic nerve disease. We have performed a meta-analysis of two independent genome-wide association studies for primary open angle glaucoma (POAG) followed by a normal-pressure glaucoma (NPG, defined by intraocular pressure (IOP) less than 22 mmHg) subgroup analysis. The single-nucleotide polymorphisms that showed the most significant associations were tested for association with a second form of glaucoma, exfoliation-syndrome glaucoma. The overall meta-analysis of the GLAUGEN and NEIGHBOR dataset results (3,146 cases and 3,487 controls) identified significant associations between two loci and POAG: the CDKN2BAS region on 9p21 (rs2157719 [G], OR = 0.69 [95%CI 0.63-0.75], p = 1.86×10⁻¹⁸), and the SIX1/SIX6 region on chromosome 14q23 (rs10483727 [A], OR = 1.32 [95%CI 1.21-1.43], p = 3.87×10⁻¹¹). In sub-group analysis two loci were significantly associated with NPG: 9p21 containing the CDKN2BAS gene (rs2157719 [G], OR = 0.58 [95% CI 0.50-0.67], p = 1.17×10⁻¹²) and a probable regulatory region on 8q22 (rs284489 [G], OR = 0.62 [95% CI 0.53-0.72], p = 8.88×10⁻¹⁰). Both NPG loci were also nominally associated with a second type of glaucoma, exfoliation syndrome glaucoma (rs2157719 [G], OR = 0.59 [95% CI 0.41-0.87], p = 0.004 and rs284489 [G], OR = 0.76 [95% CI 0.54-1.06], p = 0.021), suggesting that these loci might contribute more generally to optic nerve degeneration in glaucoma. Because both loci influence transforming growth factor beta (TGF-beta) signaling, we performed a genomic pathway analysis that showed an association between the TGF-beta pathway and NPG (permuted p = 0.009). These results suggest that neuro-protective therapies targeting TGF-beta signaling could be effective for multiple forms of glaucoma.
- Published
- 2012
- Full Text
- View/download PDF
6. Copy number variation in familial Parkinson disease.
- Author
-
Nathan Pankratz, Alexandra Dumitriu, Kurt N Hetrick, Mei Sun, Jeanne C Latourelle, Jemma B Wilk, Cheryl Halter, Kimberly F Doheny, James F Gusella, William C Nichols, Richard H Myers, Tatiana Foroud, Anita L DeStefano, and PSG-PROGENI and GenePD Investigators, Coordinators and Molecular Genetic Laboratories
- Subjects
Medicine ,Science - Abstract
Copy number variants (CNVs) are known to cause Mendelian forms of Parkinson disease (PD), most notably in SNCA and PARK2. PARK2 has a recessive mode of inheritance; however, recent evidence demonstrates that a single CNV in PARK2 (but not a single missense mutation) may increase risk for PD. We recently performed a genome-wide association study for PD that excluded individuals known to have either a LRRK2 mutation or two PARK2 mutations. Data from the Illumina370Duo arrays were re-clustered using only white individuals with high quality intensity data, and CNV calls were made using two algorithms, PennCNV and QuantiSNP. After quality assessment, the final sample included 816 cases and 856 controls. Results varied between the two CNV calling algorithms for many regions, including the PARK2 locus (genome-wide p = 0.04 for PennCNV and p = 0.13 for QuantiSNP). However, there was consistent evidence with both algorithms for two novel genes, USP32 and DOCK5 (empirical, genome-wide p-values
- Published
- 2011
- Full Text
- View/download PDF
7. Centers for Mendelian Genomics: A decade of facilitating gene discovery
- Author
-
Samantha M. Baxter, Jennifer E. Posey, Nicole J. Lake, Nara Sobreira, Jessica X. Chong, Steven Buyske, Elizabeth E. Blue, Lisa H. Chadwick, Zeynep H. Coban-Akdemir, Kimberly F. Doheny, Colleen P. Davis, Monkol Lek, Christopher Wellington, Shalini N. Jhangiani, Mark Gerstein, Richard A. Gibbs, Richard P. Lifton, Daniel G. MacArthur, Tara C. Matise, James R. Lupski, David Valle, Michael J. Bamshad, Ada Hamosh, Shrikant Mane, Deborah A. Nickerson, Heidi L. Rehm, Anne O’Donnell-Luria, Marcia Adams, François Aguet, Gulsen Akay, Peter Anderson, Corina Antonescu, Harindra M. Arachchi, Mehmed M. Atik, Christina A. Austin-Tse, Larry Babb, Tamara J. Bacus, Vahid Bahrambeigi, Suganthi Balasubramanian, Yavuz Bayram, Arthur L. Beaudet, Christine R. Beck, John W. Belmont, Jennifer E. Below, Kaya Bilguvar, Corinne D. Boehm, Eric Boerwinkle, Philip M. Boone, Sara J. Bowne, Harrison Brand, Kati J. Buckingham, Alicia B. Byrne, Daniel Calame, Ian M. Campbell, Xiaolong Cao, Claudia Carvalho, Varuna Chander, Jaime Chang, Katherine R. Chao, Ivan K. Chinn, Declan Clarke, Ryan L. Collins, Beryl Cummings, Zain Dardas, Moez Dawood, Kayla Delano, Stephanie P. DiTroia, Harshavardhan Doddapaneni, Haowei Du, Renqian Du, Ruizhi Duan, Mohammad Eldomery, Christine M. Eng, Eleina England, Emily Evangelista, Selin Everett, Jawid Fatih, Adam Felsenfeld, Laurent C. Francioli, Christian D. Frazar, Jack Fu, Emmanuel Gamarra, Tomasz Gambin, Weiniu Gan, Mira Gandhi, Vijay S. Ganesh, Kiran V. Garimella, Laura D. Gauthier, Danielle Giroux, Claudia Gonzaga-Jauregui, Julia K. Goodrich, William W. Gordon, Sean Griffith, Christopher M. Grochowski, Shen Gu, Sanna Gudmundsson, Stacey J. Hall, Adam Hansen, Tamar Harel, Arif O. Harmanci, Isabella Herman, Kurt Hetrick, Hadia Hijazi, Martha Horike-Pyne, Elvin Hsu, Jianhong Hu, Yongqing Huang, Jameson R. Hurless, Steve Jahl, Gail P. Jarvik, Yunyun Jiang, Eric Johanson, Angad Jolly, Ender Karaca, Michael Khayat, James Knight, J. Thomas Kolar, Sushant Kumar, Seema Lalani, Kristen M. Laricchia, Kathryn E. Larkin, Suzanne M. Leal, Gabrielle Lemire, Richard A. Lewis, He Li, Hua Ling, Rachel B. Lipson, Pengfei Liu, Alysia Kern Lovgren, Francesc López-Giráldez, Melissa P. MacMillan, Brian E. Mangilog, Stacy Mano, Dana Marafi, Beth Marosy, Jamie L. Marshall, Renan Martin, Colby T. Marvin, Michelle Mawhinney, Sean McGee, Daniel J. McGoldrick, Michelle Mehaffey, Betselote Mekonnen, Xiaolu Meng, Tadahiro Mitani, Christina Y. Miyake, David Mohr, Shaine Morris, Thomas E. Mullen, David R. Murdock, Mullai Murugan, Donna M. Muzny, Ben Myers, Juanita Neira, Kevin K. Nguyen, Patrick M. Nielsen, Natalie Nudelman, Emily O’Heir, Melanie C. O’Leary, Chrissie Ongaco, Jordan Orange, Ikeoluwa A. Osei-Owusu, Ingrid S. Paine, Lynn S. Pais, Justin Paschall, Karynne Patterson, Davut Pehlivan, Benjamin Pelle, Samantha Penney, Jorge Perez de Acha Chavez, Emma Pierce-Hoffman, Cecilia M. Poli, Jaya Punetha, Aparna Radhakrishnan, Matthew A. Richardson, Eliete Rodrigues, Gwendolin T. Roote, Jill A. Rosenfeld, Erica L. Ryke, Aniko Sabo, Alice Sanchez, Isabelle Schrauwen, Daryl A. Scott, Fritz Sedlazeck, Jillian Serrano, Chad A. Shaw, Tameka Shelford, Kathryn M. Shively, Moriel Singer-Berk, Joshua D. Smith, Hana Snow, Grace Snyder, Matthew Solomonson, Rachel G. Son, Xiaofei Song, Pawel Stankiewicz, Taylorlyn Stephan, V. Reid Sutton, Abigail Sveden, Diana Cornejo Sánchez, Monica Tackett, Michael Talkowski, Machiko S. Threlkeld, Grace Tiao, Miriam S. Udler, Laura Vail, Zaheer Valivullah, Elise Valkanas, Grace E. VanNoy, Qingbo S. Wang, Gao Wang, Lu Wang, Michael F. Wangler, Nicholas A. Watts, Ben Weisburd, Jeffrey M. Weiss, Marsha M. Wheeler, Janson J. White, Clara E. Williamson, Michael W. Wilson, Wojciech Wiszniewski, Marjorie A. Withers, Dane Witmer, Lauren Witzgall, Elizabeth Wohler, Monica H. Wojcik, Isaac Wong, Jordan C. Wood, Nan Wu, Jinchuan Xing, Yaping Yang, Qian Yi, Bo Yuan, Jordan E. Zeiger, Chaofan Zhang, Peng Zhang, Yan Zhang, Xiaohong Zhang, Yeting Zhang, Shifa Zhang, Huda Zoghbi, and Igna van den Veyver
- Subjects
Phenotype ,Exome Sequencing ,Humans ,Exome ,Genomics ,Article ,Genetic Association Studies ,Genetics (clinical) - Abstract
PURPOSE: Mendelian disease genomic research has undergone a massive transformation over the past decade. With increasing availability of exome and genome sequencing, the role of Mendelian research has expanded beyond data collection, sequencing, and analysis to worldwide data sharing and collaboration. METHODS: Over the past 10 years, the National Institutes of Health–supported Centers for Mendelian Genomics (CMGs) have played a major role in this research and clinical evolution. RESULTS: We highlight the cumulative gene discoveries facilitated by the program, biomedical research leveraged by the approach, and the larger impact on the research community. Beyond generating a list of gene-phenotype relationships and participating in widespread data sharing, the CMGs have created resources, tools, and training for the larger community to foster understanding of genes and genome variation. The CMGs have participated in a wide range of data sharing activities, including deposition of all eligible CMG data into the Analysis, Visualization, and Informatics Lab-space (AnVIL), sharing candidate genes through the Matchmaker Exchange and the CMG website, and sharing variants in Genotypes to Mendelian Phenotypes (Geno2MP) and VariantMatcher. CONCLUSION: The work is far from complete; strengthening communication between research and clinical realms, continued development and sharing of knowledge and tools, and improving access to richly characterized data sets are all required to diagnose the remaining molecularly undiagnosed patients.
- Published
- 2022
- Full Text
- View/download PDF
8. Genotoxic colibactin mutational signature in colorectal cancer is associated with clinicopathological features, specific genomic alterations and better survival
- Author
-
Peter Georgeson, Robert S. Steinfelder, Tabitha A. Harrison, Bernard J. Pope, Syed H. Zaidi, Conghui Qu, Yi Lin, Jihoon E. Joo, Khalid Mahmood, Mark Clendenning, Romy Walker, Elom K Aglago, Sonja I. Berndt, Hermann Brenner, Peter T. Campbell, Yin Cao, Andrew T. Chan, Jenny Chang-Claude, Niki Dimou, Kimberly F. Doheny, David A. Drew, Jane C. Figueiredo, Amy J. French, Steven Gallinger, Marios Giannakis, Graham G. Giles, Ellen L Goode, Stephen B Gruber, Andrea Gsur, Marc J. Gunter, Sophia Harlid, Michael Hoffmeister, Li Hsu, Wen-Yi Huang, Jeroen R Huyghe, JoAnn E. Manson, Victor Moreno, Neil Murphy, Rami Nassir, Christina C. Newton, Jonathan A. Nowak, Mireia Obón-Santacana, Shuji Ogino, Rish K. Pai, Nikos Papadimitrou, John D. Potter, Robert E. Schoen, Mingyang Song, Wei Sun, Amanda E. Toland, Quang M. Trinh, Kostas Tsilidis, Tomotaka Ugai, Caroline Y Um, Finlay A. Macrae, Christophe Rosty, Thomas J. Hudson, Ingrid M. Winship, Amanda I. Phipps, Mark A. Jenkins, Ulrike Peters, and Daniel D. Buchanan
- Abstract
Background and AimsThe microbiome has long been suspected of a role in colorectal cancer (CRC) tumorigenesis. The mutational signature SBS88 mechanistically links CRC development with the strain ofEscherichia coliharboring thepksisland that produces the genotoxin colibactin, but the genomic, pathological and survival characteristics associated with SBS88-positive tumors are unknown.MethodsSBS88-positive CRCs were identified from targeted sequencing data from 5,292 CRCs from 17 studies and tested for their association with clinico-pathological features, oncogenic pathways, genomic characteristics and survival.ResultsIn total, 7.5% (398/5,292) of the CRCs were SBS88-positive, of which 98.7% (392/398) were microsatellite stable/microsatellite instability low (MSS/MSI-L), compared with 80% (3916/4894) of SBS88 negative tumors (p=1.5×10-28). Analysis of MSS/MSI-L CRCs demonstrated that SBS88 positive CRCs were associated with the distal colon (OR=1.84, 95% CI=1.40-2.42, p=1×10-5) and rectum (OR=1.90, 95% CI=1.44-2.51, p=6×10-6) tumor sites compared with the proximal colon. The top seven recurrent somatic mutations associated with SBS88-positive CRCs demonstrated mutational contexts associated with colibactin-induced DNA damage, the strongest of which was theAPC:c.835-8A>G mutation (OR=65.5, 95%CI=39.0-110.0, p=3×10-80). Large copy number alterations (CNAs) including CNA loss on 14q and gains on 13q, 16q and 20p were significantly enriched in SBS88- positive CRCs. SBS88-positive CRCs were associated with better CRC-specific survival (p=0.007; hazard ratio of 0.69, 95% CI=0.52-0.90) when stratified by age, sex, study, and by stage.ConclusionSBS88-positivity, a biomarker of colibactin-induced DNA damage, can identify a novel subtype of CRC characterized by recurrent somatic mutations, copy number alterations and better survival. These findings provide new insights for treatment and prevention strategies for this subtype of CRC.
- Published
- 2023
- Full Text
- View/download PDF
9. Clonal hematopoiesis and risk of prostate cancer in large samples of European ancestry men
- Author
-
Anqi Wang, Yili Xu, Yao Yu, Kevin T Nead, TaeBeom Kim, Keren Xu, Tokhir Dadaev, Ed Saunders, Xin Sheng, Peggy Wan, Loreall Pooler, Lucy Y Xia, Stephen Chanock, Sonja I Berndt, Susan M Gapstur, Victoria Stevens, Demetrius Albanes, Stephanie J Weinstein, Vincent Gnanapragasam, Graham G Giles, Tu Nguyen-Dumont, Roger L Milne, Mark M Pomerantz, Julie A Schmidt, Konrad H Stopsack, Lorelei A Mucci, William J Catalona, Kurt N Hetrick, Kimberly F Doheny, Robert J MacInnis, Melissa C Southey, Rosalind A Eeles, Fredrik Wiklund, Zsofia Kote-Jarai, Adam J de Smith, David V Conti, Chad Huff, Christopher A Haiman, and Burcu F Darst
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
Little is known regarding the potential relationship between clonal hematopoiesis (CH) of indeterminate potential (CHIP), which is the expansion of hematopoietic stem cells with somatic mutations, and risk of prostate cancer, the fifth leading cause of cancer death of men worldwide. We evaluated the association of age-related CHIP with overall and aggressive prostate cancer risk in two large whole-exome sequencing studies of 75 047 European ancestry men, including 7663 prostate cancer cases, 2770 of which had aggressive disease, and 3266 men carrying CHIP variants. We found that CHIP, defined by over 50 CHIP genes individually and in aggregate, was not significantly associated with overall (aggregate HR = 0.93, 95% CI = 0.76–1.13, P = 0.46) or aggressive (aggregate OR = 1.14, 95% CI = 0.92–1.41, P = 0.22) prostate cancer risk. CHIP was weakly associated with genetic risk of overall prostate cancer, measured using a polygenic risk score (OR = 1.05 per unit increase, 95% CI = 1.01–1.10, P = 0.01). CHIP was not significantly associated with carrying pathogenic/likely pathogenic/deleterious variants in DNA repair genes, which have previously been found to be associated with aggressive prostate cancer. While findings from this study suggest that CHIP is likely not a risk factor for prostate cancer, it will be important to investigate other types of CH in association with prostate cancer risk.
- Published
- 2023
- Full Text
- View/download PDF
10. Variant-level matching for diagnosis and discovery: Challenges and opportunities
- Author
-
Eliete da S. Rodrigues, Sean Griffith, Renan Martin, Corina Antonescu, Jennifer E. Posey, Zeynep Coban‐Akdemir, Shalini N. Jhangiani, Kimberly F. Doheny, James R. Lupski, David Valle, Michael J. Bamshad, Ada Hamosh, Assaf Sheffer, Jessica X. Chong, Yaron Einhorn, Miro Cupak, and Nara Sobreira
- Subjects
Phenotype ,Information Dissemination ,Databases, Genetic ,Genetics ,Humans ,Exome ,Genomics ,Genetics (clinical) - Abstract
Here we describe MyGene2, Geno2MP, VariantMatcher, and Franklin; databases that provide variant-level information and phenotypic features to researchers, clinicians, healthcare providers and patients. Following the footsteps of the Matchmaker Exchange project that connects exome, genome, and phenotype databases at the gene level, these databases have as one goal to facilitate connection to one another using Data Connect, a standard for discovery and search of biomedical data from the Global Alliance for Genomics and Health (GA4GH).
- Published
- 2022
11. The impact of GeneMatcher on international data sharing and collaboration
- Author
-
Ada Hamosh, Elizabeth Wohler, Renan Martin, Sean Griffith, Eliete da S. Rodrigues, Corina Antonescu, Kimberly F. Doheny, David Valle, and Nara Sobreira
- Subjects
Rare Diseases ,Information Dissemination ,Databases, Genetic ,Genetics ,Humans ,Genetics (clinical) - Abstract
GeneMatcher (genematcher.org) is a tool designed to connect individuals with an interest in the same gene. Now used around the world to create collaborations and generate the evidence needed to support novel disease gene identification, GeneMatcher is a founding member of the Matchmaker Exchange (MME; matchmakerexchange.org) and strongest possible advocate for global data sharing including those in resource-limited environments. As of October 1, 2021, there are 12,531 submitters from 94 countries who have submitted 58,134 submissions with 13,498 unique genes in the database. Among these genes, 8970 (64%) have matched at least once and the total number of matches is 378,806, growing by about 10,000 per month. GeneMatcher submitters increase by 80-120 each month and submissions grow by800 per month, while unique genes and gene matches continue to grow steadily at rate of about 80 per month. The number of genes without a match peaked at 4371 in February of 2019 and despite the increase in the number of new submissions, the number of unique genes without a match continues to slowly decline, currently standing at 4,016. All submissions in GeneMatcher are available for matching across the MME.
- Published
- 2022
12. Deficiency of TET3 leads to a genome-wide DNA hypermethylation episignature in human whole blood
- Author
-
Trudie Cottrell, Jacques C. Giltay, Richard H. van Jaarsveld, Elles M. J. Boon, Roger E. Stevenson, Michael A. Levy, Kimberly F. Doheny, Bekim Sadikovic, G. Bradley Schaefer, Roberto Bonasio, Muhammad Ansar, Vinodh Narayanan, Mieke M. van Haelst, Jill A. Fahrner, Marleen Simon, David B. Beck, Claudia A. L. Ruivenkamp, Sivagamy Sithambaram, Teresa Romeo Luperchio, Leandros Boukas, Marie-Christine Nougues, Hannah W. Moore, Marielle Alders, Renske Oegema, M. Mahdi Motazacker, Kay Metcalfe, Cyril Mignot, Jennifer Kerkhof, Gijs W. E. Santen, Jessica Bos, Sofia Douzgou, Siddharth Banka, Keri Ramsey, Boris Keren, Human Genetics, ACS - Pulmonary hypertension & thrombosis, ANS - Complex Trait Genetics, AR&D - Amsterdam Reproduction & Development, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, and Amsterdam Reproduction & Development (AR&D)
- Subjects
Genetics ,Epigenomics ,DNA methylation ,Neurodevelopmental disorders ,Diagnostic markers ,QH426-470 ,Biology ,medicine.disease ,Phenotype ,Genome ,Article ,Neurodevelopmental disorder ,DNA demethylation ,CpG site ,medicine ,DNMT1 ,Medicine ,Epigenetics ,Author Correction ,Molecular Biology ,Genetics (clinical) - Abstract
TET3 encodes an essential dioxygenase involved in epigenetic regulation through DNA demethylation. TET3 deficiency, or Beck-Fahrner syndrome (BEFAHRS; MIM: 618798), is a recently described neurodevelopmental disorder of the DNA demethylation machinery with a nonspecific phenotype resembling other chromatin-modifying disorders, but inconsistent variant types and inheritance patterns pose diagnostic challenges. Given TET3’s direct role in regulating 5-methylcytosine and recent identification of syndrome-specific DNA methylation profiles, we analyzed genome-wide DNA methylation in whole blood of TET3-deficient individuals and identified an episignature that distinguishes affected and unaffected individuals and those with mono-allelic and bi-allelic pathogenic variants. Validation and testing of the episignature correctly categorized known TET3 variants and determined pathogenicity of variants of uncertain significance. Clinical utility was demonstrated when the episignature alone identified an affected individual from over 1000 undiagnosed cases and was confirmed upon distinguishing TET3-deficient individuals from those with 46 other disorders. The TET3-deficient signature - and the signature resulting from activating mutations in DNMT1 which normally opposes TET3 - are characterized by hypermethylation, which for BEFAHRS involves CpG sites that may be biologically relevant. This work expands the role of epi-phenotyping in molecular diagnosis and reveals genome-wide DNA methylation profiling as a quantitative, functional readout for characterization of this new biochemical category of disease.
- Published
- 2021
- Full Text
- View/download PDF
13. Author Correction: Deficiency of TET3 leads to a genome-wide DNA hypermethylation episignature in human whole blood
- Author
-
Renske Oegema, Vinodh Narayanan, Marleen Simon, Trudie Cottrell, Marie-Christine Nougues, Mieke M. van Haelst, Gijs W. E. Santen, Roger E. Stevenson, Keri Ramsey, Kay Metcalfe, Jacques C. Giltay, Sivagamy Sithambaram, Teresa Romeo Luperchio, Leandros Boukas, Marielle Alders, Hannah W. Moore, Claudia A. L. Ruivenkamp, Jessica Bos, Richard H. van Jaarsveld, Jill A. Fahrner, David B. Beck, Sofia Douzgou, Jennifer Kerkhof, Muhammad Ansar, Michael A. Levy, G. Bradley Schaefer, Siddharth Banka, Roberto Bonasio, Kimberly F. Doheny, M. Mahdi Motazacker, Cyril Mignot, Elles M. J. Boon, Boris Keren, and Bekim Sadikovic
- Subjects
Genetics ,Medicine ,Dna hypermethylation ,QH426-470 ,Biology ,Molecular Biology ,Genome ,Genetics (clinical) ,Whole blood - Published
- 2021
- Full Text
- View/download PDF
14. Identifying colorectal cancer caused by biallelic MUTYH pathogenic variants using tumor mutational signatures
- Author
-
Peter Georgeson, Tabitha A. Harrison, Bernard J. Pope, Syed H. Zaidi, Conghui Qu, Robert S. Steinfelder, Yi Lin, Jihoon E. Joo, Khalid Mahmood, Mark Clendenning, Romy Walker, Efrat L. Amitay, Sonja I. Berndt, Hermann Brenner, Peter T. Campbell, Yin Cao, Andrew T. Chan, Jenny Chang-Claude, Kimberly F. Doheny, David A. Drew, Jane C. Figueiredo, Amy J. French, Steven Gallinger, Marios Giannakis, Graham G. Giles, Andrea Gsur, Marc J. Gunter, Michael Hoffmeister, Li Hsu, Wen-Yi Huang, Paul Limburg, JoAnn E. Manson, Victor Moreno, Rami Nassir, Jonathan A. Nowak, Mireia Obón-Santacana, Shuji Ogino, Amanda I. Phipps, John D. Potter, Robert E. Schoen, Wei Sun, Amanda E. Toland, Quang M. Trinh, Tomotaka Ugai, Finlay A. Macrae, Christophe Rosty, Thomas J. Hudson, Mark A. Jenkins, Stephen N. Thibodeau, Ingrid M. Winship, Ulrike Peters, and Daniel D. Buchanan
- Subjects
Heterozygote ,Multidisciplinary ,DNA Mutational Analysis ,General Physics and Astronomy ,General Chemistry ,Colorectal cancer ,General Biochemistry, Genetics and Molecular Biology ,DNA Glycosylases ,Càncer colorectal ,Mutation ,Genetics ,Humans ,Genetic Predisposition to Disease ,Colorectal Neoplasms ,Genètica ,Germ-Line Mutation - Abstract
Carriers of germline biallelic pathogenic variants in the MUTYH gene have a high risk of colorectal cancer. We test 5649 colorectal cancers to evaluate the discriminatory potential of a tumor mutational signature specific to MUTYH for identifying biallelic carriers and classifying variants of uncertain clinical significance (VUS). Using a tumor and matched germline targeted multi-gene panel approach, our classifier identifies all biallelic MUTYH carriers and all known non-carriers in an independent test set of 3019 colorectal cancers (accuracy = 100% (95% confidence interval 99.87–100%)). All monoallelic MUTYH carriers are classified with the non-MUTYH carriers. The classifier provides evidence for a pathogenic classification for two VUS and a benign classification for five VUS. Somatic hotspot mutations KRAS p.G12C and PIK3CA p.Q546K are associated with colorectal cancers from biallelic MUTYH carriers compared with non-carriers (p = 2 × 10−23 and p = 6 × 10−11, respectively). Here, we demonstrate the potential application of mutational signatures to tumor sequencing workflows to improve the identification of biallelic MUTYH carriers.
- Published
- 2021
15. PhenoDB, GeneMatcher and VariantMatcher, tools for analysis and sharing of sequence data
- Author
-
Nara Sobreira, Kimberly F. Doheny, Shalini N. Jhangiani, Zeynep Coban-Akdemir, James R. Lupski, David Valle, Sean Griffith, Elizabeth Wohler, Eliete S. Rodrigues, Jennifer E. Posey, Corina Antonescu, Renan Paulo Martin, and Ada Hamosh
- Subjects
Computer science ,Computational biology ,DNA sequencing ,Databases, Genetic ,Genomic data ,Humans ,Pharmacology (medical) ,PhenoDB ,Exome ,Gene ,Genetics (clinical) ,Data manipulation language ,Research ,GeneMatcher ,Computational Biology ,General Medicine ,Phenotypic trait ,Genomics ,VariantMatcher ,Human genetics ,Data sharing ,ComputingMethodologies_PATTERNRECOGNITION ,Phenotype ,Medicine ,Human genome ,Software - Abstract
Background With the advent of whole exome (ES) and genome sequencing (GS) as tools for disease gene discovery, rare variant filtering, prioritization and data sharing have become essential components of the search for disease genes and variants potentially contributing to disease phenotypes. The computational storage, data manipulation, and bioinformatic interpretation of thousands to millions of variants identified in ES and GS, respectively, is a challenging task. To aid in that endeavor, we constructed PhenoDB, GeneMatcher and VariantMatcher. Results PhenoDB is an accessible, freely available, web-based platform that allows users to store, share, analyze and interpret their patients’ phenotypes and variants from ES/GS data. GeneMatcher is accessible to all stakeholders as a web-based tool developed to connect individuals (researchers, clinicians, health care providers and patients) around the globe with interest in the same gene(s), variant(s) or phenotype(s). Finally, VariantMatcher was developed to enable public sharing of variant-level data and phenotypic information from individuals sequenced as part of multiple disease gene discovery projects. Here we provide updates on PhenoDB and GeneMatcher applications and implementation and introduce VariantMatcher. Conclusion Each of these tools has facilitated worldwide data sharing and data analysis and improved our ability to connect genes to phenotypic traits. Further development of these platforms will expand variant analysis, interpretation, novel disease-gene discovery and facilitate functional annotation of the human genome for clinical genomics implementation and the precision medicine initiative.
- Published
- 2021
16. Whole genome sequencing as an investigational device for return of hereditary disease risk and pharmacogenomic results as part of the All of Us Research Program
- Author
-
Topper S, Mian S, Neben C, Donna M. Muzny, Heidi L. Rehm, Empey P, Christina M. Lockwood, Josh Smith, Stacey Gabriel, Kimberly F. Doheny, Musick A, Kimberly Walker, Eric Venner, Ginger A. Metcalf, Niall J. Lennon, Alicia Y. Zhou, Steven M. Harrison, Bradley A. Ozenberger, Deborah A. Nickerson, and Richard A. Gibbs
- Subjects
Whole genome sequencing ,Research program ,Resource (project management) ,Workflow ,Computer science ,Pharmacogenomics ,Sample (statistics) ,Observational study ,Investigational device exemption ,Data science - Abstract
The All of Us Research Program (AoURP, ‘the program’) is an initiative, sponsored by the National Institutes of Health (NIH), that aims to enroll one million people (or more) across the United States. Through repeated engagement of participants, a research resource is being created to enable a variety of future observational and interventional studies. The program has also committed to genomic data generation and returning important health-related information to participants. To do so, whole genome sequencing (WGS), variant calling processes, data interpretation, and return-of-results procedures had to be created and receive an Investigational Device Exemption (IDE) from the United States Food and Drug Administration (FDA). The performance of the entire workflow was assessed through the largest known cross-center, WGS-based, validation activity that was refined iteratively through interactions with the FDA over many months. The accuracy and precision of the WGS process as a device for the return of certain health-related genomic results was determined to be sufficient, and an IDE was granted. We present here both the process of navigating the IDE application process with the FDA and the results of the validation study as a guide to future projects which may need to follow a similar path. Future supplements to the IDE will be submitted to support additional variant classes, sample types, and any expansion to the reportable regions.
- Published
- 2021
- Full Text
- View/download PDF
17. Abstract 688: Multi-stage exome sequencing study of 17,546 aggressive and non-aggressive prostate cancer cases
- Author
-
Burcu F. Darst, Ed Saunders, Tokhir Dadaev, Xin Sheng, Peggy Wan, Loreall Pooler, Lucy Y. Xia, Stephen Chanock, Sonja I. Berndt, Susan M. Gapstur, Victoria Stevens, Demetrius Albanes, Stephanie J. Weinstein, Vincent Gnanapragasam, Graham G. Giles, Tu Nguyen-Dumont, Roger L. Milne, Mark M. Pomerantz, Julie A. Schmidt, Ruth C. Travis, Timothy J. Key, Konrad H. Stopsack, Lorelei A. Mucci, William J. Catalona, Beth Marosy, Kurt N. Hetrick, Kimberly F. Doheny, Robert J. MacInnis, Melissa C. Southey, Rosalind A. Eeles, Fredrik Wiklund, Zsofia Kote-Jarai, David V. Conti, and Christopher A. Haiman
- Subjects
Cancer Research ,Oncology - Abstract
Rare pathogenic variants in DNA repair genes have been found to influence risk of aggressive prostate cancer. We conducted a large case-only exome sequencing study to further understand the role of rare coding variation in aggressive prostate cancer in a study of 9,185 aggressive (prostate cancer death, metastatic disease, T4, or both T3 and Gleason≥8) and 8,361 non-aggressive cases (T1/T2 and Gleason≤6) of European ancestry from 19 international studies. Stage 1 samples (N=5,545) had whole exome-sequencing, and stage 2 samples (N=12,001) had targeted exome sequencing for 1,459 genes selected based on stage 1 results and previous evidence. Logistic regression models were used to evaluate gene-based tests and the aggregate effect of multiple genes to investigate whether carrying pathogenic/likely pathogenic/deleterious (P/LP/D) variants (18,759 identified) was associated with risk of aggressive prostate cancer, prostate cancer death (N=6,033), or metastatic disease (N=1,730) compared to non-aggressive disease. Gene-based tests were meta-analyzed across stages 1 and 2. BRCA2, ATM, and NBN had the most statistically significant gene-based results: BRCA2 P/LP/D variant carriers had 4.3-fold higher odds of aggressive disease (95% CI=3.15-5.86, P=4x10-20), 4.7-fold higher odds of prostate cancer death (95% CI=3.41-6.59, P=2x10-20), and 5.7-fold higher odds of metastatic disease (95% CI=3.71-8.76, P=2x10-15); ATM P/LP/D variant carriers had 2.2-fold higher odds of aggressive disease (95% CI=1.58-2.99, P=2x10-6), 2.2-fold higher odds of prostate cancer death (95% CI=1.52-3.05, P=2x10-5), and 3.0-fold higher odds of metastatic disease (95% CI=1.93-4.61, P=9x10-7); and NBN P/LP/D variant carriers had 5.9-fold higher odds of metastatic disease (95% CI=2.56-13.84, P=3x10-5). Among potentially novel genes with strong but not exome-wide significant statistical evidence were MMP19, involved in reproduction and metastasis, with carriers having 2.8-fold higher odds of prostate cancer death (95% CI=1.53-5.05, P=8x10-4); PKD2L2, involved in fertility, with carriers having 3.5-fold higher odds of prostate cancer death (95% CI=1.76-7.04, P=5x10-4); and SMPD1, involved in converting sphingomyelin to ceramide, with carriers having 5.3-fold higher odds of metastatic disease (95% CI=1.85-14.98, P=0.002). At least one P/LP/D variant within a subset of 24 previously curated candidate prostate cancer DNA repair genes was carried by 12.8% of aggressive cases (OR=1.48, 95% CI=1.34-1.64, P=3x10-14), 12.6% of cases who died due to prostate cancer (OR=1.47, 95% CI=1.31-1.65, P=3x10-11), and 15.1% of metastatic cases (OR=2.16, 95% CI=1.57-2.16, P=5x10-14) compared to 9.4% of non-aggressive cases. These findings support the importance of rare genetic variation in aggressive prostate cancer risk and may have important implications for prostate cancer risk stratification and screening. Citation Format: Burcu F. Darst, Ed Saunders, Tokhir Dadaev, Xin Sheng, Peggy Wan, Loreall Pooler, Lucy Y. Xia, Stephen Chanock, Sonja I. Berndt, Susan M. Gapstur, Victoria Stevens, Demetrius Albanes, Stephanie J. Weinstein, Vincent Gnanapragasam, Graham G. Giles, Tu Nguyen-Dumont, Roger L. Milne, Mark M. Pomerantz, Julie A. Schmidt, Ruth C. Travis, Timothy J. Key, Konrad H. Stopsack, Lorelei A. Mucci, William J. Catalona, Beth Marosy, Kurt N. Hetrick, Kimberly F. Doheny, Robert J. MacInnis, Melissa C. Southey, Rosalind A. Eeles, Fredrik Wiklund, Zsofia Kote-Jarai, David V. Conti, Christopher A. Haiman. Multi-stage exome sequencing study of 17,546 aggressive and non-aggressive prostate cancer cases [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 688.
- Published
- 2022
- Full Text
- View/download PDF
18. Germline sequencing DNA repair genes in 5,545 men with aggressive and non-aggressive prostate cancer
- Author
-
Rosalind A. Eeles, Christopher A. Haiman, David V. Conti, Xin Sheng, Zsofia Kote-Jarai, Peggy Wan, Burcu F. Darst, Kurt N. Hetrick, Demetrius Albanes, Roger L. Milne, Melissa C. Southey, Fredrik Wiklund, Lucy Xia, Lorelei A. Mucci, Loreall Pooler, Robert J. MacInnis, Graham G. Giles, Sonja I. Berndt, William J. Catalona, Vincent Gnanapragasam, Stephanie J. Weinstein, Stephen J. Chanock, Kimberly F. Doheny, Mark Pomerantz, Susan M. Gapstur, Julie A. Schmidt, Ed Saunders, Victoria L. Stevens, Tokhir Dadaev, Tu Nguyen-Dumont, Darst, Burcu F [0000-0002-6205-4632], Chanock, Stephen [0000-0002-2324-3393], Stevens, Victoria [0000-0003-0259-4407], Albanes, Demetrius [0000-0001-8330-4293], Weinstein, Stephanie J [0000-0002-3834-1535], Pomerantz, Mark [0000-0003-4914-1157], Schmidt, Julie A [0000-0002-7733-8750], Southey, Melissa C [0000-0002-6313-9005], and Apollo - University of Cambridge Repository
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,DNA Repair ,DNA repair ,PALB2 ,Genes, BRCA2 ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,P-Chloroamphetamine ,Germ-Line Mutation ,030304 developmental biology ,0303 health sciences ,business.industry ,Prostatic Neoplasms ,Cancer ,Articles ,Odds ratio ,medicine.disease ,Minor allele frequency ,Germ Cells ,030220 oncology & carcinogenesis ,business - Abstract
Background There is an urgent need to identify factors specifically associated with aggressive prostate cancer (PCa) risk. We investigated whether rare pathogenic, likely pathogenic, or deleterious (P/LP/D) germline variants in DNA repair genes are associated with aggressive PCa risk in a case-case study of aggressive vs nonaggressive disease. Methods Participants were 5545 European-ancestry men, including 2775 nonaggressive and 2770 aggressive PCa cases, which included 467 metastatic cases (16.9%). Samples were assembled from 12 international studies and germline sequenced together. Rare (minor allele frequency Results BRCA2 and PALB2 had the most statistically significant gene-based associations, with 2.5% of aggressive and 0.8% of nonaggressive cases carrying P/LP/D BRCA2 alleles (odds ratio [OR] = 3.19, 95% confidence interval [CI] = 1.94 to 5.25, P = 8.58 × 10-7) and 0.65% of aggressive and 0.11% of nonaggressive cases carrying P/LP/D PALB2 alleles (OR = 6.31, 95% CI = 1.83 to 21.68, P = 4.79 × 10-4). ATM had a nominal association, with 1.6% of aggressive and 0.8% of nonaggressive cases carrying P/LP/D ATM alleles (OR = 1.88, 95% CI = 1.10 to 3.22, P = .02). In aggregate, P/LP/D alleles within 24 literature-curated candidate PCa DNA repair genes were more common in aggressive than nonaggressive cases (carrier frequencies = 14.2% vs 10.6%, respectively; P = 5.56 × 10-5). However, this difference was non-statistically significant (P = .18) on excluding BRCA2, PALB2, and ATM. Among these 24 genes, P/LP/D carriers had a 1.06-year younger diagnosis age (95% CI = -1.65 to 0.48, P = 3.71 × 10-4). Conclusions Risk conveyed by DNA repair genes is largely driven by rare P/LP/D alleles within BRCA2, PALB2, and ATM. These findings support the importance of these genes in both screening and disease management considerations.
- Published
- 2020
19. Discovery of common and rare genetic risk variants for colorectal cancer
- Author
-
Charles M. Connolly, Deborah A. Nickerson, Jian Gong, Sébastien Küry, Barbara Pardini, Brent W. Zanke, Andrea Gsur, Jochen Hampe, Coral Arnau-Collell, M. Henar Alonso, Elio Riboli, Annika Lindblom, Ulrike Peters, Gad Rennert, Tabitha A. Harrison, Lori C. Sakoda, Caroline McNeil, Flavio Lejbkowicz, Hyun Min Kang, David J. Hunter, Martha L. Slattery, Miguel Rodríguez-Barranco, Christina Bamia, Satu Männistö, Timothy J. Key, W. James Gauderman, Gonçalo R. Abecasis, Sanford D. Markowitz, Laurence N. Kolonel, Mark A. Jenkins, Yi Lin, Robin Myte, Hedy S. Rennert, Neil Murphy, Antonia Trichopoulou, Christopher I. Li, Ross L. Prentice, Sai Chen, Stephanie J. Weinstein, Kristin E. Anderson, Hua Ling, Mitul Shah, Philipp Hofer, Wen Yi Huang, Sergi Castellví-Bel, Susanna C. Larsson, Maria Dolores Chirlaque, Wei Zheng, Stephanie L. Schmit, Cecelia A. Laurie, Soo-Chin Lee, David Forman, Andrea N. Burnett-Hartman, Giovanna Masala, Sarah C. Nelson, Michael O. Woods, Charles Kooperberg, Qing Zhang, Sonja I. Berndt, Christopher S. Carlson, Katja Butterbach, Hyeong Rok Kim, Rebecca D. Jackson, David Van Den Berg, Michael C. Bassik, Amanda J. Cross, Sushma S. Thomas, Clemens Schafmayer, Anna H. Wu, Douglas F. Easton, Robert W. Haile, Ludmila Vodickova, Graham G. Giles, Yu Ru Su, Jenny Chang-Claude, Lorena Moreno, Peter C. Scacheri, Stefanie Brezina, Min-Ho Shin, Steven Gallinger, Bethany Van Guelpen, Daniel D. Buchanan, Roger L. Milne, Stephen J. Chanock, Tin Louie, Tameka Shelford, Emily White, Kala Visvanathan, Loic Le Marchand, Veronika Vymetalkova, Roxann G. Ingersoll, Temitope O. Keku, Stephanie A. Bien, Fredrick R. Schumacher, Wan-Ling Hsu, Amanda E. Toland, John S. Grove, Noralane M. Lindor, Faye Elliott, Leon Raskin, Heather Hampel, Joshua D. Smith, Vicente Martín, David V. Conti, Sjoerd G. Elias, Henk J. van Kranen, Manish Gala, Daniela Seminara, Syed H.E. Zaidi, Suzanne M. Leal, Tilman Kühn, Korbinian Weigl, Marc J. Gunter, Cornelia M. Ulrich, Peyton Greenside, Victor Moreno, John D. Potter, Michael Hoffmeister, Eric J. Jacobs, Catherine M. Tangen, Jihyoun Jeon, Fränzel J.B. Van Duijnhoven, Andrew T. Chan, Stephen B. Gruber, John A. Baron, Alicja Wolk, Edith J. M. Feskens, Demetrius Albanes, Amit Joshi, Bette J. Caan, Polly A. Newcomb, Stéphane Bézieau, Elizabeth M. Gillanders, Anshul Kundaje, Elizabeth A. Platz, Michael Wainberg, Sun-Seog Kweon, C. Roland Wolf, Gemma Ibáñez-Sanz, Shuji Ogino, Emiko Kobayashi, Richard B. Hayes, Patrick S. Parfrey, Katarina Cuk, Stephen N. Thibodeau, Kenneth Offit, David Duggan, Sophia Harlid, Pavel Vodicka, Juergen Boehm, Christa Stegmaier, Jeroen R. Huyghe, Joseph Vijai, Sang-Hee Cho, Elizabeth W. Pugh, Rachel Pearlman, Alessio Naccarati, Marilena Melas, Graham Casey, Jane Romm, Stephan Buch, Phyllis J. Goodman, Albert de la Chapelle, John L. Hopper, Zsofia K. Stadler, Corinne E. Joshu, Liesel M. FitzGerald, Wolfgang Lieb, Aung Ko Win, Keith R. Curtis, Hermann Brenner, Christopher K. Edlund, Li Hsu, Conghui Qu, Peter T. Campbell, Robert E. Schoen, Heiner Boeing, D. Timothy Bishop, Kimberly F. Doheny, Sabina Sieri, Barbara L. Banbury, Mathieu Lemire, Jane C. Figueiredo, Gregory Idos, Katerina Shulman, Thomas J. Hudson, Melissa C. Southey, Duncan C. Thomas, Paul D.P. Pharoah, Mila Pinchev, Vittorio Perduca, Rocky Fischer, Volker Arndt, William M. Grady, Nasa Sinnott-Armstrong, N. Charlotte Onland-Moret, David M. Levine, Li Li, Dallas R. English, Health Research Board - Ireland, Department of Medical Genetics, HMNC Brain Health, Case Western Reserve University [Cleveland], National Cancer Institute, NIH, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), WHO Collaborating Center for Food and Nutrition Policies, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), Service de Génétique, Centre hospitalier universitaire de Nantes (CHU Nantes), Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Division of Cancer Epidemiology, Cancer Genome Project, The Wellcome Trust Sanger Institute [Cambridge], Division of Signaling Biology, Ontario Cancer Institute, Consorcio de Investigación Biomédica en Red especializado en Epidemiología y Salud Pública (CIBERESP), Los Centros de Investigación Biomédica en Red (CIBER), National Institute of Standards and Technology [Gaithersburg] (NIST), Ohio State University [Columbus] (OSU), Centre for Cancer Genetic Epidemiology, University of Cambridge [UK] (CAM), Section of Epidemiology and Biostatistics, Leeds Institute of Molecular Medicine, Cancer Epidemiology Centre, Cancer Council Victoria, Division of Human Nutrition, Wageningen University and Research [Wageningen] (WUR), Division of Public Health Sciences, Fred Hutchinson Cancer Research Center [Seattle] (FHCRC), Northern and Yorkshire Cancer Registry and Information Service, Familial Gastrointestinal Cancer Registry, Mount Sinai Hospital [Toronto, Canada] (MSH), University of Melbourne, Nutrition and Metabolism Section, International Agency for Cancer Research (IACR), Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, London School of Hygiene and Tropical Medicine (LSHTM), Fraunhofer Institute for Manufacturing Engineering and Automation (Fraunhofer IPA), Fraunhofer (Fraunhofer-Gesellschaft), Centre for Molecular , Environmental, Genetic and Analytic (MEGA) Epidemiology, University of Melbourne-Centre for Molecular, Melbourne School of Population Health, Department of Mathematics, University of Warwick, Warwick Mathematics Institute (WMI), University of Warwick [Coventry]-University of Warwick [Coventry], Department of Statistics, Penn State University, University of Pennsylvania [Philadelphia], Tata Memorial Centre, Cancer Epidemiology Unit, University of Oxford [Oxford], Thermo Fisher Scientific, Thermo Fisher Scientific Inc., Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Baylor University-Baylor University, National University Health System, Department of Community Medicine and Epidemiology, CHS National Cancer Control Center, Ontario Institute for Cancer Research [Canada] (OICR), Ontario Institute for Cancer Research, Laboratoire de Génie Electrique de Grenoble (G2ELab), Institut polytechnique de Grenoble - Grenoble Institute of Technology (Grenoble INP )-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Chronic Disease Epidemiology and Prevention Unit, National Institute for Health and Welfare [Helsinki], Dell-EMC, Molecular and Nutritional Epidemiology Unit (ISPO), Cancer Research and Prevention Institute, Clinical Genetics Service, Memorial Sloane Kettering Cancer Center [New York], Department of Pathology, Brigham and Women's Hospital [Boston], University Medical Center [Utrecht], Mathématiques Appliquées Paris 5 (MAP5 - UMR 8145), Université Paris Descartes - Paris 5 (UPD5)-Institut National des Sciences Mathématiques et de leurs Interactions (INSMI)-Centre National de la Recherche Scientifique (CNRS), Department of Oncology, Department of Epidemiology and Biostatistics, Imperial College London, Department of Visceral and Thoracic Surgery [Kiel, Germany], University Hospital Schleswig-Holstein [Kiel, Germany], Nutritional Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, entre for Molecular, Environmental, Genetic and Analytic (MEGA) Epidemiology, Department of Laboratory Medicine and Department of Pathology, Mayo Clinic College of Medicine, Department of Molecular Virology, Immunology and Medical Genetics [Colombus], Ohio State University [Columbus] (OSU)-College of Medicine and Public Health [Colombus], Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School [Athens], Department of Medical Biosciences and Pathology, Umeå University, Institute of Experimental Medicine, Czech Academy of Sciences [Prague] (CAS), Dundee Technopole, CXR Biosciences Ltd, Karolinska Institutet [Stockholm], CINTRA / SEEE Nanyang Technological University, Nanyang Technological University [Singapour], Center for Astrophysical Sciences [Baltimore], Johns Hopkins University (JHU), Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology (MIT), Department of Preventive Medicine, University of Southern California (USC), Biomedical Research Centre Network for Rare Diseases, CIBER de Enfermedades Raras (CIBERER), Medstar Research Institute, Department of Genome Sciences [Seattle] (GS), University of Washington [Seattle], Department of Internal Medicine, Epidemiology, Human Genetics, University of Michigan [Ann Arbor], University of Michigan System-University of Michigan System, Wageningen University and Research Centre [Wageningen] (WUR), Mount Sinai Hospital (MSH), Fraunhofer Institute for Manufacturing Engineering and Automation [Stuttgart] (IPA), Université Joseph Fourier - Grenoble 1 (UJF)-Institut polytechnique de Grenoble - Grenoble Institute of Technology (Grenoble INP)-Institut Polytechnique de Grenoble - Grenoble Institute of Technology-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA), Czech Academy of Sciences [Prague] (ASCR), Huyghe, Jeroen R [0000-0001-6027-9806], Harrison, Tabitha A [0000-0002-4173-7530], Chen, Sai [0000-0003-3106-5643], Schmit, Stephanie L [0000-0001-5931-1194], Jeon, Jihyoun [0000-0001-7003-3412], Schumacher, Fredrick R [0000-0002-3073-7463], Nelson, Sarah C [0000-0002-2109-6465], Sinnott-Armstrong, Nasa A [0000-0003-4490-0601], Alonso, M Henar [0000-0003-0285-5451], Arndt, Volker [0000-0001-9320-8684], Bézieau, Stéphane [0000-0003-0095-1319], Bishop, D Timothy [0000-0002-8752-8785], Brezina, Stefanie [0000-0001-5238-6900], Buchanan, Daniel D [0000-0003-2225-6675], Chanock, Stephen J [0000-0002-2324-3393], de la Chapelle, Albert [0000-0001-9345-9248], Easton, Douglas F [0000-0003-2444-3247], Hampe, Jochen [0000-0002-2421-6127], Hayes, Richard B [0000-0002-0918-661X], Hofer, Philipp [0000-0003-2550-6019], Huang, Wen-Yi [0000-0002-4440-3368], Hudson, Thomas J [0000-0002-1376-4849], Jacobs, Eric J [0000-0002-8458-7659], Jenkins, Mark A [0000-0002-8964-6160], Joshi, Amit D [0000-0001-7581-6934], Küry, Sébastien [0000-0001-5497-0465], Larsson, Susanna C [0000-0003-0118-0341], Laurie, Cecelia A [0000-0001-6569-2501], Martín, Vicente [0000-0003-0552-2804], Masala, Giovanna [0000-0002-5758-9069], Milne, Roger L [0000-0001-5764-7268], Naccarati, Alessio [0000-0001-5774-0905], Newcomb, Polly A [0000-0001-8786-0043], Pardini, Barbara [0000-0001-9571-4257], Perduca, Vittorio [0000-0003-0339-0473], Pharoah, Paul DP [0000-0001-8494-732X], Raskin, Leon [0000-0003-1195-7214], Rennert, Gad [0000-0002-8512-068X], Shin, Min-Ho [0000-0002-2217-5624], Toland, Amanda E [0000-0002-0271-1792], Vijai, Joseph [0000-0002-7933-151X], Weigl, Korbinian [0000-0003-4453-2036], Win, Aung Ko [0000-0002-2794-5261], Wolk, Alicja [0000-0001-7387-6845], Zheng, Wei [0000-0003-1226-070X], Bassik, Michael C [0000-0001-5185-8427], Moreno, Victor [0000-0002-2818-5487], Peters, Ulrike [0000-0001-5666-9318], and Apollo - University of Cambridge Repository
- Subjects
Male ,Nutrition and Disease ,Colorectal cancer ,IDENTIFIES 6 ,Genome-wide association study ,0302 clinical medicine ,Risk Factors ,Voeding en Ziekte ,Genotype ,ComputingMilieux_MISCELLANEOUS ,Avaluació del risc per la salut ,Genetics & Heredity ,Genetics ,0303 health sciences ,COLON-CANCER ,11 Medical And Health Sciences ,Middle Aged ,3. Good health ,Medical genetics ,Female ,RNA, Long Noncoding ,Colorectal Neoplasms ,Life Sciences & Biomedicine ,Signal Transduction ,EXPRESSION ,medicine.medical_specialty ,SUSCEPTIBILITY LOCI ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Polymorphism, Single Nucleotide ,Article ,Health risk assessment ,03 medical and health sciences ,QUALITY-CONTROL ,Càncer colorectal ,Journal Article ,medicine ,Life Science ,Humans ,Genetic Predisposition to Disease ,GENOME-WIDE ASSOCIATION ,METAANALYSIS ,VLAG ,Aged ,030304 developmental biology ,Global Nutrition ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Wereldvoeding ,Science & Technology ,ORGAN SIZE ,MUTATIONS ,Haplotype ,Case-control study ,06 Biological Sciences ,medicine.disease ,Genetic architecture ,Case-Control Studies ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,Human genome ,LYSOPHOSPHATIDIC ACID ,030217 neurology & neurosurgery ,Developmental Biology ,Genome-Wide Association Study - Abstract
To further dissect the genetic architecture of colorectal cancer (CRC), we performed whole-genome sequencing of 1,439 cases and 720 controls, imputed discovered sequence variants and Haplotype Reference Consortium panel variants into genome-wide association study data, and tested for association in 34,869 cases and 29,051 controls. Findings were followed up in an additional 23,262 cases and 38,296 controls. We discovered a strongly protective 0.3% frequency variant signal at CHD1. In a combined meta-analysis of 125,478 individuals, we identified 40 new independent signals at P, Reporting Summary. Further information on experimental design is available in the Life Sciences Reporting Summary linked to this article.
- Published
- 2018
- Full Text
- View/download PDF
20. Association of Smoking, Alcohol Consumption, Blood Pressure, Body Mass Index, and Glycemic Risk Factors With Age-Related Macular Degeneration
- Author
-
Itay Chowers, Stacy M Meuer, R. Theodore Smith, Bamini Gopinath, Brendan J Vote, Thierry Léveillard, David A Mackey, Dwight Stambolian, Jamie E Craig, José-Alain Sahel, David J Hunter, Michael L Klein, Jane Romm, Robyn H Guymer, Mingyao Li, J. L. Haines, Emily L. Moore, J Allie McGrath, Chloe M. Stanton, Danni Lin, Jessica N Cooke Bailey, Anton Orlin, Anita Agarwal, Frank G Holz, Debra A Schaumberg, Valerie Kuan, Christine A. Curcio, Ken Flagg, Sudha K Iyengar, Sebanti Sengupta, Bal Dhillon, Joanna E. Merriam, Janette Hall, Bernhard H F Weber, Caroline Brandl, Donald Zack, Eric Souied, Yara T. E. Lechanteur, Christina A Rennie, Mathias Gorski, Murray H Brilliant, Denise J. Morgan, Barbara Truitt, Daniel E Weeks, Thomas Langmann, Aroon D. Hingorani, Gerald Liew, Andrea J Richardson, Neal S Peachey, John Blangero, Alasdair Warwick, Humma Shahid, Eiko K de Jong, Kari E Branham, S. V. Goverdhan, Paul Mitchell, Angela J Cree, Margaux A. Morrison, Rebecca J Sardell, Ian J Constable, Michael A. Hauser, Zhenglin Yang, Reneé Laux, G. Rudolph, David Cho, Jie Jin Wang, Albert Caramoy, Jaclyn L Kovach, Alexander Brucker, Frédéric Blond, Hongrong Luo, Michael B Gorin, Robert P Igo, Caroline C W Klaver, Lebriz Ersoy, Timothy Isaacs, Adnan Tufail, Gabriëlle H.S. Buitendijk, Nicholas Katsanis, Stephen Burgess, Carel B Hoyng, Reecha Sofat, Ivana K Kim, Mohammad Othman, Ian L McAllister, Giuliana Silvestri, Helena Hai Liang, Margaret DeAngelis, Matthew P Johnson, Ava G Tan, Felix Grassmann, Lindsay A Farrer, Alex W Hewitt, Hong Ouyang, Cindy Wen, Henry Ferreyra, Milam A Brantley, Melinda Cain, Caroline Hayward, Kristine E. Lee, Linn Gieser, Isabelle Audo, Evangelia E Tsironi, Nicole T.M. Saksens, Hendrik P N Scholl, Stephen G Schwartz, Matthias Olden, Saddek Mohand-Said, Scott J Hebbring, Joshua D Hoffman, Shira Hagbi-Levi, Anthony T Moore, Mustapha Benchaboune, Lars G Fritsche, Margaret A Pericak-Vance, Iris M Heid, Kyu Hyung Park, Jennifer L Bragg-Gresham, Hélène Blanché, Alexis Boleda, Rando Allikmets, John R Heckenlively, Kathryn P Burdon, Elisa Bala, Rinki Ratnapriya, Kimberly F Doheny, Xiaowei Zhan, Sascha Fauser, Claudia N von Strachwitz, Ronald Klein, Johanna R. Foerster, Wilmar Igl, Andrew J Lotery, Klaus Stark, Matthew Brooks, Jane C Khan, Emily Y Chew, Paul N Baird, Cornelia M Van Duijn, Chelsea E. Myers, Anneke I den Hollander, Monique D Courtenay, Zhiguang Su, Yingda Jiang, William K Scott, Tammy M Martin, Armin Wolf, Jeeyun Ahn, John C. Merriam, Eric A Postel, Guanping Mao, Emmanuelle Souzeau, Barbara E K Klein, Terrie Kitchner, Stewart Lake, Anand Swaroop, Valentina Cipriani, Tina Schick, Stephanie A. Hagstrom, Alan M. Kwong, Daniel Chen, Gonçalo R. Abecasis, Matthew Schu, Michelle Grunin, John R.W. Yates, Peter Campochiaro, Kang Zhang, and Jean-François Deleuze
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Alcohol Drinking ,medicine.medical_treatment ,Visual Acuity ,Angiogenesis Inhibitors ,Blood Pressure ,Type 2 diabetes ,Blindness ,Lower risk ,Body Mass Index ,Risk Factors ,Internal medicine ,Mendelian randomization ,Humans ,Medicine ,Risk factor ,Glycemic ,business.industry ,Smoking ,Odds ratio ,Mendelian Randomization Analysis ,medicine.disease ,Ophthalmology ,Diabetes Mellitus, Type 2 ,Wet Macular Degeneration ,Smoking cessation ,business ,Body mass index ,Genome-Wide Association Study - Abstract
Importance Advanced age-related macular degeneration (AMD) is a leading cause of blindness in Western countries. Causal, modifiable risk factors need to be identified to develop preventive measures for advanced AMD. Objective To assess whether smoking, alcohol consumption, blood pressure, body mass index, and glycemic traits are associated with increased risk of advanced AMD. Design, Setting, Participants This study used 2-sample mendelian randomization. Genetic instruments composed of variants associated with risk factors at genome-wide significance (P
- Published
- 2021
- Full Text
- View/download PDF
21. Patients with a Kabuki syndrome phenotype demonstrate DNA methylation abnormalities
- Author
-
Débora Romeo Bertola, Chrissie M. Ongaco, Kimberly F. Doheny, Vera Ayres Meloni, Maria Isabel Melaragno, David Valle, Jane Romm, Nara Sobreira, Chong Ae Kim, Li Zhang, Regina Célia Mingroni-Netto, Martha Brucato, Hans T. Bjornsson, Christine Ladd-Acosta, and Ana Beatriz Alvarez Perez
- Subjects
Male ,0301 basic medicine ,030105 genetics & heredity ,Article ,03 medical and health sciences ,Loss of Function Mutation ,Genotype ,Genetics ,medicine ,Humans ,Missense mutation ,Abnormalities, Multiple ,Epigenetics ,Child ,Genetics (clinical) ,Loss function ,biology ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,medicine.disease ,Hematologic Diseases ,Phenotype ,030104 developmental biology ,KMT2A ,Histone ,Vestibular Diseases ,Case-Control Studies ,Face ,DNA methylation ,biology.protein ,Female ,Kabuki syndrome ,Myeloid-Lymphoid Leukemia Protein - Abstract
Kabuki syndrome is a monogenic disorder caused by loss of function variants in either of two genes encoding histone-modifying enzymes. We performed targeted sequencing in a cohort of 27 probands with a clinical diagnosis of Kabuki syndrome. Of these, 12 had causative variants in the two known Kabuki syndrome genes. In 2, we identified presumptive loss of function de novo variants in KMT2A (missense and splice site variants), a gene that encodes another histone modifying enzyme previously exclusively associated with Wiedermann-Steiner syndrome. Although Kabuki syndrome is a disorder of histone modification, we also find alterations in DNA methylation among individuals with a Kabuki syndrome diagnosis relative to matched normal controls, regardless of whether they carry a variant in KMT2A or KMT2D or not. Furthermore, we observed characteristic global abnormalities of DNA methylation that distinguished patients with a loss of function variant in KMT2D or missense or splice site variants in either KMT2D or KMT2A from normal controls. Our results provide new insights into the relationship of genotype to epigenotype and phenotype and indicate cross-talk between histone and DNA methylation machineries exposed by inborn errors of the epigenetic apparatus.
- Published
- 2017
- Full Text
- View/download PDF
22. Concordance between Research Sequencing and Clinical Pharmacogenetic Genotyping in the eMERGE-PGx Study
- Author
-
Laura J. Rasmussen-Torvik, Kimberly F. Doheny, Kejian Zhang, Stuart A. Scott, Ammar Husami, Michael D. Linderman, Sarah C. Stallings, Adam S. Gordon, Iftikhar J. Kullo, Robert R. Freimuth, Marylyn D. Ritchie, Wendy A. Wolf, Teri A. Manolio, Jared B. Hawkins, Lynn Ivacic, Berta Almoguera, Cynthia A. Prows, Renata Pellegrino, Hakon Hakonarson, Maureen E. Smith, and Aniwaa Owusu Obeng
- Subjects
0301 basic medicine ,Genotype ,Genotyping Techniques ,Concordance ,Computational biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Medicine ,Genotyping ,Alleles ,Genetics ,Polymorphism, Genetic ,business.industry ,High-Throughput Nucleotide Sequencing ,Regular Article ,Sequence Analysis, DNA ,Pharmacogenomic Testing ,030104 developmental biology ,Pharmacogenetics ,030220 oncology & carcinogenesis ,Research studies ,Molecular Medicine ,business - Abstract
There has been extensive debate about both the necessity of orthogonal confirmation of next-generation sequencing (NGS) results in Clinical Laboratory Improvement Amendments-approved laboratories and return of research NGS results to participants enrolled in research studies. In eMERGE-PGx, subjects underwent research NGS using PGRNseq and orthogonal targeted genotyping in clinical laboratories, which prompted a comparison of genotyping results between platforms. Concordance (percentage agreement) was reported for 4077 samples tested across nine combinations of research and clinical laboratories. Retesting was possible on a subset of 1792 samples, and local laboratory directors determined sources of genotype discrepancy. Research NGS and orthogonal clinical genotyping had an overall per sample concordance rate of 0.972 and per variant concordance rate of 0.997. Genotype discrepancies attributed to research NGS were because of sample switching (preanalytical errors), whereas the majority of genotype discrepancies (92.3%) attributed to clinical genotyping were because of allele dropout as a result of rare variants interfering with primer hybridization (analytical errors). These results highlight the analytical quality of clinically significant pharmacogenetic variants derived from NGS and reveal important areas for research and clinical laboratories to address with quality management programs.
- Published
- 2017
- Full Text
- View/download PDF
23. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer
- Author
-
Harald Surowy, Rulla M. Tamimi, Javier Benitez, Wing-Yee Lo, Celine M. Vachon, Nadege Presneau, John J. Spinelli, Ann Smeets, Hoda Anton-Culver, Veli-Matti Kosma, Christopher A. Haiman, Martha J. Shrubsole, Ross L. Prentice, Diana Eccles, Ursula Eilber, Loic Le Marchand, Katri Pylkäs, Jirong Long, Michael P. Lux, Sara Margolin, Hedy S. Rennert, Tom Maishman, Mary B. Daly, Rita K. Schmutzler, Julian Peto, Sune F. Nielsen, Eric Hahnen, Niclas Håkansson, Børge G. Nordestgaard, Mitul Shah, Matthias W. Beckmann, Anthony J. Swerdlow, Barbara Burwinkel, Rudolf Kaaks, Usha Menon, William J. Tapper, Argyrios Ziogas, Peter Hillemanns, Fares Al-Ejeh, Roger L. Milne, Wolfgang Janni, Pascal Guénel, Mikael Eriksson, Clarice R. Weinberg, Kyriaki Michailidou, Jonathan Beesley, Marike Gabrielson, J. Esteban Castelao, Margriet Collée, Janet E. Olson, Gad Rennert, Per Broberg, Rob A. E. M. Tollenaar, David Cox, Paolo Peterlongo, Helian Feng, Brian D. Carter, Nichola Johnson, Emmanouil Saloustros, Dijana Plaseska-Karanfilska, Kimberly F. Doheny, Paul L. Auer, Hans Wildiers, Jacques Simard, Michael Untch, Per Hall, Martine Dumont, Julie M. Cunningham, Thilo Dörk, Mary Beth Terry, Jenny Chang-Claude, Lang Wu, Irene L. Andrulis, Xiaohong R. Yang, Caroline Baynes, Isabel dos-Santos-Silva, Douglas F. Easton, Wei Shi, Emily Hallberg, Camilla Wendt, Hiltrud Brauch, Diana Torres, Olufunmilayo I. Olopade, David Van Den Berg, Georgia Chenevix-Trench, Alfons Meindl, Stig E. Bojesen, Jonine D. Figueroa, Dale P. Sandler, Vessela N. Kristensen, Christine L. Clarke, Wei Zheng, Manuela Gago-Dominguez, E Rozali, Henrik Flyger, Sheila Seal, Guanmengqian Huang, Marjanka K. Schmidt, Håkan Olsson, Christopher G. Scott, Kamila Czene, Laura Fachal, Rodney J. Scott, Jennifer Stone, Sara Y. Brucker, Qin Wang, David J. Hunter, Maya Ghoussaini, Christoph Engel, Keith Humphreys, Susan M. Gapstur, Daniel C. Tessier, Paolo Radice, John L. Hopper, Audrey Y. Jung, Lucy Xia, Atocha Romero, Chenjie Zeng, Peter A. Fasching, Jan Lubinski, Anna González-Neira, Nazneen Rahman, Robert N. Hoover, Thérèse Truong, Fergus J. Couch, Anna Marie Mulligan, Robert J. MacInnis, Ute Hamann, Hanne Meijers-Heijboer, Brigitte Rack, Simon S. Cross, Federico Canzian, J.-P. Meyer, Sara Lindström, Natalia Bogdanova, Trinidad Caldés, Olivia Fletcher, Peter Kraft, Elinor J. Sawyer, Alexander Gusev, Louise A. Brinton, Diether Lambrechts, Bingshan Li, Kristan J. Aronson, Jane Romm, Anja Rudolph, Peter Devilee, Qiuyin Cai, Arto Mannermaa, Elad Ziv, Alice S. Whittemore, Abigail Thomas, Hermann Brenner, Montserrat Garcia-Closas, Patrick Neven, Kristine Jones, Miriam Dwek, Sibylle Loibl, Heli Nevanlinna, Jolanta Lissowska, Susan L. Neuhausen, Elza Khusnutdinova, Marina Bermisheva, Alicja Wolk, Lin Fritschi, Xingyi Guo, Angela Cox, Michael Jones, Xiaoqing Chen, Esther M. John, Richard Barfield, Volker Arndt, Patricia Harrington, Quinten Waisfisz, Daniel Vincent, Antoinette Hollestelle, Dimitrios Mavroudis, JoAnn E. Manson, Joe Dennis, Walter C. Willett, Stacey L. Edwards, Melissa C. Southey, Andreas Schneeweiss, Jack A. Taylor, Robert Winqvist, Mia M. Gaudet, David E. Goldgar, Anna Jakubowska, Paul D.P. Pharoah, Kathrin Thöne, Dieter Flesch-Janys, Mark S. Goldberg, Craig Luccarini, Sten Cornelissen, Jingmei Li, Michael J. Kerin, Myrto Barrdahl, Xiao-Ou Shu, Alison M. Dunning, Manjeet K. Bolla, Carl Blomqvist, Graham G. Giles, Hans Christiansen, A. Heather Eliassen, Valerie Rhenius, Alexander Hein, Belynda Hicks, Ivana Maleva Kostovska, Tongguang Cheng, Yingchang Lu, CCA - Cancer biology and immunology, Amsterdam Neuroscience - Complex Trait Genetics, Human genetics, Amsterdam Reproduction & Development (AR&D), Clinical Genetics, Medical Oncology, Guo, Xingyi [0000-0001-5269-1294], Al-Ejeh, Fares [0000-0002-1553-0077], Li, Bingshan [0000-0003-2129-168X], Gusev, Alexander [0000-0002-7980-4620], Andrulis, Irene L [0000-0002-4226-6435], Arndt, Volker [0000-0001-9320-8684], Brauch, Hiltrud [0000-0001-7531-2736], Collée, Margriet [0000-0002-9272-9346], Cox, Angela [0000-0002-5138-1099], Cunningham, Julie M [0000-0002-8159-3025], Fachal, Laura [0000-0002-7256-9752], Fletcher, Olivia [0000-0001-9387-7116], Hein, Alexander [0000-0003-2601-3398], Hicks, Belynda [0000-0001-8014-4888], Hollestelle, Antoinette [0000-0003-1166-1966], Jakubowska, Anna [0000-0002-5650-0501], Khusnutdinova, Elza [0000-0003-2987-3334], Li, Jingmei [0000-0001-8587-7511], Menon, Usha [0000-0003-3708-1732], Nevanlinna, Heli [0000-0002-0916-2976], Nordestgaard, Børge G [0000-0002-1954-7220], Pylkäs, Katri [0000-0002-2449-0521], Rennert, Gad [0000-0002-8512-068X], Romero, Atocha [0000-0002-1634-7397], Saloustros, Emmanouil [0000-0002-0485-0120], Scott, Christopher G [0000-0003-1340-0647], Shrubsole, Martha J [0000-0002-5591-7575], Wolk, Alicja [0000-0001-7387-6845], Ziogas, Argyrios [0000-0003-4529-3727], Pharoah, Paul DP [0000-0001-8494-732X], Milne, Roger L [0000-0001-5764-7268], Easton, Douglas F [0000-0003-2444-3247], Zheng, Wei [0000-0003-1226-070X], Apollo - University of Cambridge Repository, Human Genetics, and ARD - Amsterdam Reproduction and Development
- Subjects
0301 basic medicine ,Risk ,medicine.medical_specialty ,Gene Expression ,Genome-wide association study ,Breast Neoplasms ,Biology ,Genome-wide association studies ,Polymorphism, Single Nucleotide ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,SDG 3 - Good Health and Well-being ,Genetic model ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic association ,medicine.disease ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Expression quantitative trait loci ,Medical genetics ,Female ,Gene expression ,Breast Cancer Genetics ,Genome-Wide Association Study - Abstract
The breast cancer risk variants identified in genome-wide association studies explain only a small fraction of the familial relative risk, and the genes responsible for these associations remain largely unknown. To identify novel risk loci and likely causal genes, we performed a transcriptome-wide association study evaluating associations of genetically predicted gene expression with breast cancer risk in 122,977 cases and 105,974 controls of European ancestry. We used data from the Genotype-Tissue Expression Project to establish genetic models to predict gene expression in breast tissue and evaluated model performance using data from The Cancer Genome Atlas. Of the 8,597 genes evaluated, significant associations were identified for 48 at a Bonferroni-corrected threshold of P −6, including 14 genes at loci not yet reported for breast cancer. We silenced 13 genes and showed an effect for 11 on cell proliferation and/or colony-forming efficiency. Our study provides new insights into breast cancer genetics and biology.
- Published
- 2019
- Full Text
- View/download PDF
24. Insights into genetics, human biology and disease gleaned from family based genomic studies
- Author
-
Deborah A. Nickerson, Pengfei Liu, Nara Sobreira, Jessica X. Chong, Eric Boerwinkle, Davut Pehlivan, Samantha Baxter, Nan Wu, V. Reid Sutton, David Valle, Jill A. Rosenfeld, Dimitri Avramopoulos, Tamar Harel, Anne H. O’Donnell-Luria, Murat Gunel, Jennifer E. Posey, Tara C. Matise, Richard P. Lifton, James R. Lupski, Heidi L. Rehm, Donna M. Muzny, Claudia M.B. Carvalho, Steven Buyske, Zeynep Coban Akdemir, Daniel G. MacArthur, C. D. Boehm, Mark Gerstein, Kimberly F. Doheny, Janson White, Richard A. Gibbs, Sushant Kumar, Shalini N. Jhangiani, Michael J. Bamshad, Shrikant Mane, P. Dane Witmer, and Ada Hamosh
- Subjects
0301 basic medicine ,Genomics ,Locus (genetics) ,Computational biology ,030105 genetics & heredity ,Biology ,Article ,03 medical and health sciences ,symbols.namesake ,Genetic Heterogeneity ,Databases, Genetic ,Exome Sequencing ,Humans ,Genetic Predisposition to Disease ,Allele ,Genetics (clinical) ,Exome sequencing ,Genome, Human ,Genetic Diseases, Inborn ,Oligogenic Inheritance ,Human genetics ,United States ,Pedigree ,030104 developmental biology ,National Institutes of Health (U.S.) ,Mendelian inheritance ,symbols ,Human genome - Abstract
Identifying genes and variants contributing to rare disease phenotypes and Mendelian conditions informs biology and medicine, yet potential phenotypic consequences for variation of >75% of the ~20,000 annotated genes in the human genome are lacking. Technical advances to assess rare variation genome-wide, particularly exome sequencing (ES), enabled establishment in the United States of the National Institutes of Health (NIH)-supported Centers for Mendelian Genomics (CMGs) and have facilitated collaborative studies resulting in novel “disease gene” discoveries. Pedigree-based genomic studies and rare variant analyses in families with suspected Mendelian conditions have led to the elucidation of hundreds of novel disease genes and highlighted the impact of de novo mutational events, somatic variation underlying nononcologic traits, incompletely penetrant alleles, phenotypes with high locus heterogeneity, and multilocus pathogenic variation. Herein, we highlight CMG collaborative discoveries that have contributed to understanding both rare and common diseases and discuss opportunities for future discovery in single-locus Mendelian disorder genomics. Phenotypic annotation of all human genes; development of bioinformatic tools and analytic methods; exploration of non-Mendelian modes of inheritance including reduced penetrance, multilocus variation, and oligogenic inheritance; construction of allelic series at a locus; enhanced data sharing worldwide; and integration with clinical genomics are explored. Realizing the full contribution of rare disease research to functional annotation of the human genome, and further illuminating human biology and health, will lay the foundation for the Precision Medicine Initiative.
- Published
- 2019
25. Multi-Omics Analysis Reveals a HIF Network and Hub Gene EPAS1 Associated with Lung Adenocarcinoma
- Author
-
Claudio W. Pikielny, Kimberly F. Doheny, Jolanta Lissowska, James McKay, Beth A. Marosy, Geoffrey Liu, Vladimir Janout, Rayjean J. Hung, Ciprian Bolca, Kurt N. Hetrick, Xiangjun Xiao, Zhaoxi Wang, Ghislaine Scelo, Milica Kontic, Xihong Lin, David C. Christiani, John K. Field, David Zaridze, Beata Swiatkowska, Paul Brennan, Ruyang Zhang, Yongyue Wei, Elizabeth K. Pugh, Stephanie M. Gogarten, Li Su, Christopher I. Amos, Cathy C. Laurie, and Nofer Institute of Occupational Medicine
- Subjects
0301 basic medicine ,Lung adenocarcinoma ,Male ,Integrated analysis ,Lung Neoplasms ,Gene regulatory network ,lcsh:Medicine ,Adenocarcinoma of Lung ,Computational biology ,Biology ,Adenocarcinoma ,Hypoxia-inducible factor ,Genome ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Non-small cell lung cancer ,Carcinoma, Non-Small-Cell Lung ,Basic Helix-Loop-Helix Transcription Factors ,Gene family ,Humans ,Genetic Predisposition to Disease ,Gene ,Genetic Association Studies ,Aged ,lcsh:R5-920 ,lcsh:R ,General Medicine ,Epigenome ,DNA Methylation ,Middle Aged ,Hypoxia-Inducible Factor 1, alpha Subunit ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,DNA methylation ,Human genome ,Network analysis ,Female ,lcsh:Medicine (General) ,Research Paper - Abstract
Recent technological advancements have permitted high-throughput measurement of the human genome, epigenome, metabolome, transcriptome, and proteome at the population level. We hypothesized that subsets of genes identified from omic studies might have closely related biological functions and thus might interact directly at the network level. Therefore, we conducted an integrative analysis of multi-omic datasets of non-small cell lung cancer (NSCLC) to search for association patterns beyond the genome and transcriptome. A large, complex, and robust gene network containing well-known lung cancer-related genes, including EGFR and TERT, was identified from combined gene lists for lung adenocarcinoma. Members of the hypoxia-inducible factor (HIF) gene family were at the center of this network. Subsequent sequencing of network hub genes within a subset of samples from the Transdisciplinary Research in Cancer of the Lung-International Lung Cancer Consortium (TRICL-ILCCO) consortium revealed a SNP (rs12614710) in EPAS1 associated with NSCLC that reached genome-wide significance (OR = 1.50; 95% CI: 1.31–1.72; p = 7.75 × 10−9). Using imputed data, we found that this SNP remained significant in the entire TRICL-ILCCO consortium (p = .03). Additional functional studies are warranted to better understand interrelationships among genetic polymorphisms, DNA methylation status, and EPAS1 expression., • Previous Evidence • As a major participant of Transdisciplinary Research in Cancer of the Lung-International Lung Cancer Consortium (TRICL-ILCCO), we continued monitoring the progress of genomic and other omics studies of lung cancer through regular communications within the consortium and literature search using the PubMed database. The TRICL-ILCCO is one of the largest international consortium of lung cancer which includes many major ongoing lung cancer case-control and cohort studies with the aim of sharing comparable data. Although GWAS successfully defined many lung cancer-associated genomic loci, the integrative studies of multi-dimensional high throughput “-omics” measurements from tumor tissues and corresponding blood specimens are limited. • Added value of this study • We identified a HIFs-EGFR-HDAC4-TERT network associated with lung adenocarcinoma, and subsequent sequencing of network hub genes identified a new locus within EPAS1 that is associated with lung cancer risk. This locus is in hub gene EPAS1, which is a key member of the HIF family involved in every aspect of cancer development and progression. • Implications of all the available evidence • We developed a network building approach for the integrative analysis of multi-omic datasets. The integration of multi-dimensional high throughput “-omics” measurements from tumor tissues and corresponding blood specimens, together with new systems strategies for diagnostics, enables the identification of cancer biomarkers that will facilitate pre-symptomatic diagnosis, stratification of disease, assessment of disease progression, evaluation of patient response to therapy, and identification of recurrences.
- Published
- 2018
26. Rare, low frequency and common coding variants in CHRNA5 and their contribution to nicotine dependence in European and African Americans
- Author
-
Nora Franceschini, Oscar H. Franco, Laura J. Bierut, John P. Rice, Kenneth Rice, Sarah Bertelsen, Donna K. Arnett, Michael R. Brown, Lynda M. Rose, J. C. Wang, Louis Fox, A.G. Uitterlinden, George D. Wilson, B. Marosy, Lei Chen, Nancy L. Saccone, Alanna C. Morrison, R. G. Barr, Dina Vojinovic, Bruce M. Psaty, Najaf Amin, A. Hofman, Jerry A. Stitzel, Eric O. Johnson, Kurt N. Hetrick, Karen Schwander, Rob Culverhouse, Traci M. Bartz, Ervin R. Fox, Cathy C. Laurie, Jie Yao, Sharon L.R. Kardia, Dorothy K. Hatsukami, Ursula M. Schick, Wei Zhao, Mary F. Feitosa, C M van Duijn, Alison Goate, Megan L. Grove, Naomi Breslau, Alexander P. Reiner, Sarah M. Hartz, Stephanie M. Gogarten, Steven M. Foltz, Paul M. Ridker, Xiuqing Guo, Erin B. Ware, Daniel I. Chasman, Dabeeru C. Rao, Solomon K. Musani, Emily Olfson, Kimberly F. Doheny, Ingrid B. Borecki, Epidemiology, and Internal Medicine
- Subjects
Adult ,Male ,0301 basic medicine ,Fagerstrom Test for Nicotine Dependence ,Nonsynonymous substitution ,Nerve Tissue Proteins ,Receptors, Nicotinic ,White People ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Genetic variation ,Humans ,Medicine ,Genetic Predisposition to Disease ,Molecular Biology ,Exome ,Genetics ,biology ,business.industry ,CHRNA5 ,Genetic Variation ,Tobacco Use Disorder ,Odds ratio ,Middle Aged ,Black or African American ,Minor allele frequency ,Psychiatry and Mental health ,030104 developmental biology ,Meta-analysis ,biology.protein ,Female ,business ,030217 neurology & neurosurgery - Abstract
The common nonsynonymous variant rs16969968 in the α5 nicotinic receptor subunit gene (CHRNA5) is the strongest genetic risk factor for nicotine dependence in European Americans and contributes to risk in African Americans. To comprehensively examine whether other CHRNA5 coding variation influences nicotine dependence risk, we performed targeted sequencing on 1582 nicotine-dependent cases (Fagerstrom Test for Nicotine Dependence score⩾4) and 1238 non-dependent controls, with independent replication of common and low frequency variants using 12 studies with exome chip data. Nicotine dependence was examined using logistic regression with individual common variants (minor allele frequency (MAF)⩾0.05), aggregate low frequency variants (0.05>MAF⩾0.005) and aggregate rare variants (MAF
- Published
- 2015
- Full Text
- View/download PDF
27. The Genetic Basis of Mendelian Phenotypes: Discoveries, Challenges, and Opportunities
- Author
-
Francesc López-Giráldez, Eric Boerwinkle, Julie Hoover-Fong, Alan F. Scott, Frederic Reinier, V. Reid Sutton, Kimberly F. Doheny, Holly K. Tabor, Tomasz Gambin, Murat Gunel, Debra J. H. Mathews, Hua Ling, Aravinda Chakravarti, Donna M. Muzny, Richard A. Gibbs, Elizabeth Blue, Karynne E. Patterson, Suzanne M. Leal, Michael J. Bamshad, Richard P. Lifton, Shrikant Mane, Wojciech Wiszniewski, James R. Lupski, Nara Sobreira, C. D. Boehm, Joshua D. Smith, Lee Watkins, P. Dane Witmer, Kaya Bilguvar, Zeynep Coban Akdemir, Jessica X. Chong, Shalini N. Jhangiani, Jay Shendure, Kurt N. Hetrick, Kati J. Buckingham, Ada Hamosh, Martin Kircher, Deborah A. Nickerson, David Valle, Dimitri Avramopoulos, Margaret J. McMillin, and Tanya M. Harrell
- Subjects
Genetics ,Biomedical knowledge ,Genetics, Medical ,Genetic Diseases, Inborn ,Proteins ,Genomics ,Review ,Disease ,Biology ,Phenotype ,3. Good health ,symbols.namesake ,Genotype-phenotype distinction ,Mendelian inheritance ,symbols ,Humans ,Causal link ,Genetics(clinical) ,Gene ,Genetics (clinical) - Abstract
Discovering the genetic basis of a Mendelian phenotype establishes a causal link between genotype and phenotype, making possible carrier and population screening and direct diagnosis. Such discoveries also contribute to our knowledge of gene function, gene regulation, development, and biological mechanisms that can be used for developing new therapeutics. As of February 2015, 2,937 genes underlying 4,163 Mendelian phenotypes have been discovered, but the genes underlying ∼50% (i.e., 3,152) of all known Mendelian phenotypes are still unknown, and many more Mendelian conditions have yet to be recognized. This is a formidable gap in biomedical knowledge. Accordingly, in December 2011, the NIH established the Centers for Mendelian Genomics (CMGs) to provide the collaborative framework and infrastructure necessary for undertaking large-scale whole-exome sequencing and discovery of the genetic variants responsible for Mendelian phenotypes. In partnership with 529 investigators from 261 institutions in 36 countries, the CMGs assessed 18,863 samples from 8,838 families representing 579 known and 470 novel Mendelian phenotypes as of January 2015. This collaborative effort has identified 956 genes, including 375 not previously associated with human health, that underlie a Mendelian phenotype. These results provide insight into study design and analytical strategies, identify novel mechanisms of disease, and reveal the extensive clinical variability of Mendelian phenotypes. Discovering the gene underlying every Mendelian phenotype will require tackling challenges such as worldwide ascertainment and phenotypic characterization of families affected by Mendelian conditions, improvement in sequencing and analytical techniques, and pervasive sharing of phenotypic and genomic data among researchers, clinicians, and families.
- Published
- 2015
- Full Text
- View/download PDF
28. Biology‐Driven Gene‐Gene Interaction Analysis of Age‐Related Cataract in the eMERGE Network
- Author
-
Anastasia Lucas, Molly A. Hall, John Connolly, Sarah A. Pendergrass, Richard L. Berg, Gail P. Jarvik, Marylyn D. Ritchie, Catherine A. McCarty, Eric B. Larson, Helena Kuivaniemi, Terrie Kitchner, Tamara R. Vrabec, David R. Crosslin, John B. Harley, John M. Wallace, Jonathan L. Haines, Gerard Tromp, Kimberly F. Doheny, Dana C. Crawford, Mariza de Andrade, Shefali S. Verma, and David Carrell
- Subjects
Male ,Epidemiology ,Genomics ,Single-nucleotide polymorphism ,Biology ,Logistic regression ,Polymorphism, Single Nucleotide ,Cataract ,Population Groups ,Gene interaction ,genetic interaction ,Cell Adhesion ,Electronic Health Records ,Humans ,SNP ,Gene ,Research Articles ,Genetics (clinical) ,Genetics ,Models, Genetic ,complex disease ,association ,Age Factors ,Intron ,Computational Biology ,Middle Aged ,3. Good health ,Case-Control Studies ,Data Interpretation, Statistical ,Multiple comparisons problem ,Female ,Gene-Environment Interaction ,Software ,Genome-Wide Association Study ,Signal Transduction - Abstract
Bioinformatics approaches to examine gene-gene models provide a means to discover interactions between multiple genes that underlie complex disease. Extensive computational demands and adjusting for multiple testing make uncovering genetic interactions a challenge. Here, we address these issues using our knowledge-driven filtering method, Biofilter, to identify putative single nucleotide polymorphism (SNP) interaction models for cataract susceptibility, thereby reducing the number of models for analysis. Models were evaluated in 3,377 European Americans (1,185 controls, 2,192 cases) from the Marshfield Clinic, a study site of the Electronic Medical Records and Genomics (eMERGE) Network, using logistic regression. All statistically significant models from the Marshfield Clinic were then evaluated in an independent dataset of 4,311 individuals (742 controls, 3,569 cases), using independent samples from additional study sites in the eMERGE Network: Mayo Clinic, Group Health/University of Washington, Vanderbilt University Medical Center, and Geisinger Health System. Eighty-three SNP-SNP models replicated in the independent dataset at likelihood ratio test P < 0.05. Among the most significant replicating models was rs12597188 (intron of CDH1)–rs11564445 (intron of CTNNB1). These genes are known to be involved in processes that include: cell-to-cell adhesion signaling, cell-cell junction organization, and cell-cell communication. Further Biofilter analysis of all replicating models revealed a number of common functions among the genes harboring the 83 replicating SNP-SNP models, which included signal transduction and PI3K-Akt signaling pathway. These findings demonstrate the utility of Biofilter as a biology-driven method, applicable for any genome-wide association study dataset.
- Published
- 2015
- Full Text
- View/download PDF
29. The PAGE Study: How Genetic Diversity Improves Our Understanding of the Architecture of Complex Traits
- Author
-
Steve Buyske, Claudia Schurmann, Andrés Moreno-Estrada, Cathy C. Laurie, Ulrike Peters, Yesha Patel, Eimear E. Kenny, Michael Preuss, Hannah Poisner, Kari E. North, Carlos Bustamante, Christopher A. Haiman, Stephanie A. Bien, Anne E. Justice, Katherine K. Nishimura, Yuqing Li, Loic Le Marchand, Unhee Lim, E Stahl, Christopher R. Gignoux, Alexandra Sockell, Paul Norman, Ewa Deelman, Yingchang Lu, Rebecca D. Jackson, Huckins Lm, Gerardo Heiss, Kimberly F. Doheny, Chani J. Hodonsky, Brenna M. Henn, Benyam Hailu, Genevieve L. Wojcik, Jane Romm, Misa Graff, Loos R, Christy L. Avery, Danyu Lin, José Luis Ambite, Kris Young, Eric Boerwinkle, Kathleen C. Barnes, Gillian M. Belbin, Bridget M Lin, Lindsay Fernández-Rhodes, Cecilia A. Laurie, Christian Caberto, Sung-Hyuk Park, Tara C. Matise, Abhishek Vishnu, Loreall Pooler, Ran Tao, Elena P. Sorokin, Ron Do, Lucia A. Hindorff, Sarah C. Nelson, Heather M. Highland, L R Wilkens, Cheryl A. Winkler, Iona Cheng, Acuna-Alonso, Matthew P. Conomos, Myriam Fornage, Xin Sheng, Girish N. Nadkarni, Christopher S. Carlson, Ruth H. Walker, Janina M. Jeff, Jeffrey Haessler, Daniel O. Stram, Sabati C, Sachiko Yoneyama, Jonathan M. Kocarnik, Christina L. Wassel, Timothy A. Thornton, Charles Kooperberg, Veronica Wendy Setiawan, Yao Hu, Sinead Cullina, Canizales-Quinteroes S, Niha Zubair, Melissa A. Richard, Karan Vahi, Karla Sandoval, Marie Verbanck, Erwin P. Bottinger, and Alexander P. Reiner
- Subjects
0303 health sciences ,medicine.medical_specialty ,education.field_of_study ,Genetic diversity ,Public health ,Population ,Genomics ,Genome-wide association study ,Disease ,Biology ,Health equity ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Evolutionary biology ,030220 oncology & carcinogenesis ,medicine ,education ,030304 developmental biology ,Genetic association - Abstract
Summary/AbstractGenome-wide association studies (GWAS) have laid the foundation for investigations into the biology of complex traits, drug development, and clinical guidelines. However, the dominance of European-ancestry populations in GWAS creates a biased view of the role of human variation in disease, and hinders the equitable translation of genetic associations into clinical and public health applications. The Population Architecture using Genomics and Epidemiology (PAGE) study conducted a GWAS of 26 clinical and behavioral phenotypes in 49,839 non-European individuals. Using strategies designed for analysis of multi-ethnic and admixed populations, we confirm 574 GWAS catalog variants across these traits, and find 38 secondary signals in known loci and 27 novel loci. Our data shows strong evidence of effect-size heterogeneity across ancestries for published GWAS associations, substantial benefits for fine-mapping using diverse cohorts, and insights into clinical implications. We strongly advocate for continued, large genome-wide efforts in diverse populations to reduce health disparities.
- Published
- 2017
- Full Text
- View/download PDF
30. Genetic analyses of diverse populations improves discovery for complex traits
- Author
-
Paul Norman, Mariaelisa Graff, Elena P. Sorokin, Michael Preuss, Ron Do, Victor Acuña-Alonso, Lucia A. Hindorff, Yingchang Lu, Eli A. Stahl, Ruth J. F. Loos, Steven Buyske, Tara C. Matise, José Luis Ambite, Lindsay Fernández-Rhodes, Abhishek Vishnu, Matthew P. Conomos, Chiara Sabatti, Katherine K. Nishimura, Myriam Fornage, Xin Sheng, Samuel Canizales-Quinteros, Carlos Bustamante, Christopher R. Gignoux, Girish N. Nadkarni, Christopher S. Carlson, Daniel O. Stram, Sachi Yoneyama, Christian Caberto, Stephanie A. Bien, Gerardo Heiss, Iona Cheng, Janina M. Jeff, Timothy A. Thornton, Charles Kooperberg, Lynne R. Wilkens, Alexandra Sockell, Loreall Pooler, Ryan W. Walker, Cheryl A. Winkler, Christy L. Avery, Ran Tao, Rebecca D. Jackson, Heather M. Highland, Kathleen C. Barnes, Sungshim L. Park, Yesha Patel, Christina L. Wassel, Sinead Cullina, Jonathan M. Kocarnik, Claudia Schurmann, Eimear E. Kenny, Jeffrey Haessler, Yuqing Li, Cathy C. Laurie, Benyam Hailu, Kristin L. Young, Jane Romm, Danyu Lin, Brenna M. Henn, Gillian M. Belbin, Anne E. Justice, Alexander P. Reiner, Bridget M Lin, Yao Hu, Cecelia A. Laurie, Niha Zubair, Veronica Wendy Setiawan, Andrés Moreno-Estrada, Melissa A. Richard, Kari E. North, Karan Vahi, Christopher A. Haiman, Loic Le Marchand, Ulrike Peters, Chani J. Hodonsky, Eric Boerwinkle, Laura M. Huckins, Sarah C. Nelson, Hannah Poisner, Unhee Lim, Ewa Deelman, Genevieve L. Wojcik, Kimberly F. Doheny, Karla Sandoval, Marie Verbanck, and Erwin P. Bottinger
- Subjects
0301 basic medicine ,Asian Continental Ancestry Group ,Male ,Multifactorial Inheritance ,General Science & Technology ,Genetics, Medical ,Population ,Black People ,Genomics ,Genome-wide association study ,Disease ,Biology ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Asian People ,2.5 Research design and methodologies (aetiology) ,Medical ,Genetics ,Humans ,Genetic Testing ,education ,Minority Groups ,Genetic association ,African Continental Ancestry Group ,education.field_of_study ,Multidisciplinary ,Health Equity ,Prevention ,Human Genome ,Health Status Disparities ,Hispanic or Latino ,Blacks ,Precision medicine ,Genetic architecture ,Health equity ,Body Height ,United States ,Asians ,030104 developmental biology ,Evolutionary biology ,Women's Health ,Female ,Hispanic Americans ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Genome-wide association studies (GWAS) have laid the foundation for investigations into the biology of complex traits, drug development and clinical guidelines. However, the majority of discovery efforts are based on data from populations of European ancestry1–3. In light of the differential genetic architecture that is known to exist between populations, bias in representation can exacerbate existing disease and healthcare disparities. Critical variants may be missed if they have a low frequency or are completely absent in European populations, especially as the field shifts its attention towards rare variants, which are more likely to be population-specific4–10. Additionally, effect sizes and their derived risk prediction scores derived in one population may not accurately extrapolate to other populations11,12. Here we demonstrate the value of diverse, multi-ethnic participants in large-scale genomic studies. The Population Architecture using Genomics and Epidemiology (PAGE) study conducted a GWAS of 26 clinical and behavioural phenotypes in 49,839 non-European individuals. Using strategies tailored for analysis of multi-ethnic and admixed populations, we describe a framework for analysing diverse populations, identify 27 novel loci and 38 secondary signals at known loci, as well as replicate 1,444 GWAS catalogue associations across these traits. Our data show evidence of effect-size heterogeneity across ancestries for published GWAS associations, substantial benefits for fine-mapping using diverse cohorts and insights into clinical implications. In the United States—where minority populations have a disproportionately higher burden of chronic conditions13—the lack of representation of diverse populations in genetic research will result in inequitable access to precision medicine for those with the highest burden of disease. We strongly advocate for continued, large genome-wide efforts in diverse populations to maximize genetic discovery and reduce health disparities. © 2019, The Author(s), under exclusive licence to Springer Nature Limited.
- Published
- 2017
31. Imputation-Based Genomic Coverage Assessments of Current Genotyping Arrays: Illumina HumanCore, OmniExpress, Multi-Ethnic global array and sub-arrays, Global Screening Array, Omni2.5M, Omni5M, and Affymetrix UK Biobank
- Author
-
Jane Romm, Elizabeth W. Pugh, Kimberly F. Doheny, Cathy C. Laurie, and Sarah C. Nelson
- Subjects
Genetics ,education.field_of_study ,Linkage disequilibrium ,Population ,Genetic variants ,Human genome ,Computational biology ,Biology ,1000 Genomes Project ,education ,Genotyping ,Biobank ,Imputation (genetics) - Abstract
Genotyping arrays have been widely adopted as an efficient means to interrogate variation across the human genome. Genetic variants may be observed either directly, via genotyping, or indirectly, through linkage disequilibrium with a genotyped variant. The total proportion of genomic variation captured by an array, either directly or indirectly, is referred to as “genomic coverage.” Here we use genotype imputation and Phase 3 of the 1000 Genomes Project to assess genomic coverage of several modern genotyping arrays. We find that in general, coverage increases with increasing array density. However, arrays designed to cover specific populations may yield better coverage in those populations compared to denser arrays not tailored to the given population. Ultimately, array choice involves trade-offs between cost, density, and coverage, and our work helps inform investigators weighing these choices and trade-offs.
- Published
- 2017
- Full Text
- View/download PDF
32. Genome-Wide Association Analysis Reveals Genetic Heterogeneity of Sjögren's Syndrome According to Ancestry
- Author
-
Mi Y. Lam, Hisanori Umehara, Caroline H. Shiboski, Troy E. Daniels, Quenna Wong, Kimberly F. Doheny, David M. Levine, Frederick B. Vivino, Yan Zhao, Hector Lanfranchi, Muthiah Srinivasan, Morten Schiødt, Caitlin P. McHugh, Stephen Challacombe, Cathy C. Laurie, John S. Greenspan, Alan N. Baer, Kimberly E. Taylor, Stephen Shiboski, and Lindsey A. Criswell
- Subjects
0301 basic medicine ,Male ,Genome-wide association study ,Salivary Glands ,Major Histocompatibility Complex ,0302 clinical medicine ,Gene Frequency ,Polymorphism (computer science) ,Immunologic ,Lectins ,Genotype ,Receptors ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Registries ,Aetiology ,Receptors, Immunologic ,Genetics ,C-Type ,Sjogrens Syndrome ,Adaptor Proteins ,Single Nucleotide ,STAT4 Transcription Factor ,3. Good health ,Sjogren's Syndrome ,Phenotype ,Interferon Regulatory Factors ,Public Health and Health Services ,Female ,Asian Continental Ancestry Group ,Clinical Sciences ,Immunology ,European Continental Ancestry Group ,Biology ,Salivary Glands, Minor ,Autoimmune Disease ,Polymorphism, Single Nucleotide ,White People ,Article ,03 medical and health sciences ,Genetic Heterogeneity ,Asian People ,Rheumatology ,Clinical Research ,Genome-Wide Association Analysis ,Humans ,Lectins, C-Type ,Genetic Predisposition to Disease ,Polymorphism ,Allele frequency ,Genotyping ,Adaptor Proteins, Signal Transducing ,Autoantibodies ,030203 arthritis & rheumatology ,Genetic heterogeneity ,Human Genome ,Case-control study ,Signal Transducing ,Arthritis & Rheumatology ,Minor ,030104 developmental biology ,Case-Control Studies ,Trans-Activators ,Digestive Diseases ,Genome-Wide Association Study - Abstract
Author(s): Taylor, Kimberly E; Wong, Quenna; Levine, David M; McHugh, Caitlin; Laurie, Cathy; Doheny, Kimberly; Lam, Mi Y; Baer, Alan N; Challacombe, Stephen; Lanfranchi, Hector; Schiodt, Morten; Srinivasan, M; Umehara, Hisanori; Vivino, Frederick B; Zhao, Yan; Shiboski, Stephen C; Daniels, Troy E; Greenspan, John S; Shiboski, Caroline H; Criswell, Lindsey A | Abstract: ObjectiveThe Sjogren's International Collaborative Clinical Alliance (SICCA) is an international data registry and biorepository derived from a multisite observational study of participants in whom genotyping was performed on the Omni2.5M platform and who had undergone deep phenotyping using common protocol-directed methods. The aim of this study was to examine the genetic etiology of Sjogren's syndrome (SS) across ancestry and disease subsets.MethodsWe performed genome-wide association study analyses using SICCA subjects and external controls obtained from dbGaP data sets, one using all participants (1,405 cases, 1,622 SICCA controls, and 3,125 external controls), one using European participants (585, 966, and 580, respectively), and one using Asian participants (460, 224, and 901, respectively) with ancestry adjustments via principal components analyses. We also investigated whether subphenotype distributions differ by ethnicity, and whether this contributes to the heterogeneity of genetic associations.ResultsWe observed significant associations in established regions of the major histocompatibility complex (MHC), IRF5, and STAT4 (P = 3 × 10-42 , P = 3 × 10-14 , and P = 9 × 10-10 , respectively), and several novel suggestive regions (those with 2 or more associations at P l 1 × 10-5 ). Two regions have been previously implicated in autoimmune disease: KLRG1 (P = 6 × 10-7 [Asian cluster]) and SH2D2A (P = 2 × 10-6 [all participants]). We observed striking differences between the associations in Europeans and Asians, with high heterogeneity especially in the MHC; representative single-nucleotide polymorphisms from established and suggestive regions had highly significant differences in the allele frequencies in the study populations. We showed that SSA/SSB autoantibody production and the labial salivary gland focus score criteria were associated with the first worldwide principal component, indicative of higher non-European ancestry (P = 4 × 10-15 and P = 4 × 10-5 , respectively), but that subphenotype differences did not explain most of the ancestry differences in genetic associations.ConclusionGenetic associations with SS differ markedly according to ancestry; however, this is not explained by differences in subphenotypes.
- Published
- 2017
- Full Text
- View/download PDF
33. Identification of 12 new susceptibility loci for different histotypes of epithelial ovarian cancer
- Author
-
Eitan Friedman, Mattias Van Heetvelde, Jennifer B. Permuth, Joseph Vijai, Patricia A. Ganz, Jennifer A. Doherty, Argyrios Ziogas, Bernard Peissel, Edwin S. Iversen, Angela Brooks-Wilson, Ingo B. Runnebaum, Amanda B. Spurdle, Heli Nevanlinna, Kenneth Offit, Laura Papi, Georgia Chenevix-Trench, Saundra S. Buys, Martin Köbel, Fabienne Lesueur, Elizabeth W. Pugh, Joe Dennis, Sylvie Mazoyer, Diana Eccles, Shirley Hodgson, Jolanta Lissowska, Judy Garber, Pascal Pujol, Kristin K. Zorn, Orland Diez, Marcia Adams, Thomas Conner, Renée T. Fortner, Tjoung-Won Park-Simon, Vanesa García-Barberán, Kerstin Rhiem, Norbert Arnold, Karoline Kuchenbaecker, Susan M. Domchek, María Josefa Mosteiro García, Matthias W. Beckmann, Alex Henderson, Melissa C. Larson, Jane Romm, Anja Rudolph, Steven A. Narod, Mats Jernetz, Jolanta Kupryjanczyk, Natalia Bogdanova, Jacob Musinsky, Helga B. Salvesen, Jonathan Beesley, Paolo Peterlongo, Arif B. Ekici, Clarice R. Weinberg, Marion Piedmonte, Christian F. Singer, Robert L. Nussbaum, Katja K.H. Aben, Michael J. Birrer, Juul T. Wijnen, Elizabeth M. Poole, Phuong L. Mai, David J. Hunter, Tanja Pejovic, Athanassios Vratimos, Barbara Wappenschmidt, Nicolas Wentzensen, Marcus Q. Bernardini, Leigha Senter, Terence Cescon, Daniel W. Cramer, Silvia Tognazzo, Drakoulis Yannoukakos, Jacopo Azzollini, Ignace Vergote, Karen H. Lu, Gustavo C. Rodriguez, Julian Adlard, Tomasz Huzarski, Mark H. Greene, Susan L. Neuhausen, Marina Bermisheva, Alicja Wolk, Paulo C Lyra, Usha Menon, Ralf Bützow, Siddhartha Kar, Manuel R. Teixeira, Conxi Lázaro, Agnieszka Dansonka-Mieszkowska, Aleksandra Gentry-Maharaj, Zsofia K. Stadler, Melissa C. Southey, Ramunas Janavicius, Douglas F. Easton, Digna R. Velez Edwards, Jocelyne Chiquette, Karin Kast, Jonathan Tyrer, Georg Pfeiler, Tara M. Friebel, Bruno Buecher, Goska Leslie, Jackie Cook, Catherine M. Phelan, Steve Ellis, Estrid Høgdall, Beth Y. Karlan, Anthony J. Swerdlow, Sarah E. Ferguson, Rosalind Glasspool, Frans B. L. Hogervorst, Lotte Nedergaard, Britton Trabert, Jack A. Taylor, Irene L. Andrulis, Paolo Radice, Dennis J. Hazelett, Mads Thomassen, Dong Liang, Joseph H. Rothstein, Loren Lipworth, Melissa A. Merritt, Ana Vega, Petra H.M. Peeters, Claus Høgdall, Anna M. Piskorz, Bernardo Bonanni, Janet M. Lee, Malcolm C. Pike, Clemens Liebrich, Zachary C. Fogarty, Bent Ejlertsen, Yuan Chun Ding, Dieter Niederacher, Michael E. Carney, Dominique Stoppa-Lyonnet, Nadine Tung, Curtis Olswold, Ana Osorio, Fiona Bruinsma, Christine Walsh, Fabienne Prieur, Lara E. Sucheston-Campbell, Stephen B. Gruber, Maartje J. Hooning, George Fountzilas, Amanda Black, David E. Goldgar, Anna Jakubowska, Paul D.P. Pharoah, Angela R. Bradbury, Helene Holland, Ruth C. Travis, Susana Banerjee, Penelope M. Webb, Brooke L. Fridley, Clara Bodelon, Mary Anne Rossing, Yen Y. Tan, Rosa B. Barkardottir, Jong Won Lee, Stephen J. Chanock, Bruce Poppe, Sandra Fert Ferrer, Melissa Moffitt, Taymaa May, Gustavo Mendoza-Fandiño, Christopher A. Haiman, Alicia Beeghly-Fadiel, Rebecca Sutphen, Michelle A.T. Hildebrandt, Lambertus A. Kiemeney, Thilo Dörk, Douglas A. Levine, Gerasimos Aravantinos, Celeste Leigh Pearce, Sue K. Park, David Van Den Berg, Louise Izatt, Hannah P. Yang, Graham G. Giles, Linda S. Cook, John R. McLaughlin, Nick Orr, Weiva Sieh, Raymonda Varon-Mateeva, Marco Montagna, Honglin Song, Laura Ottini, Ruea-Yea Huang, Joanna Moes-Sosnowska, Anders Bojesen, David M. O'Malley, Andrew K. Godwin, Lucy Side, Sung-Won Kim, Lukasz Szafron, Christoph Engel, Harvey A. Risch, Alexander Hein, Penny Soucy, Elza Khusnutdinova, Ana Peixoto, Arto Leminen, Cora M. Aalfs, Matthias Dürst, Mary B. Daly, Patricia Rice, Nadeem Siddiqui, Dale P. Sandler, Ava Kwong, Madalene Earp, Marjorie J. Riggan, Inge Søkilde Pedersen, Susanne K. Kjaer, Mercedes Durán, Joellen M. Schildkraut, James D. Brenton, D. Gareth Evans, Liisa M. Pelttari, Kimberly F. Doheny, Karen Hosking, Miquel Angel Pujana, Salina B. Chan, Joan Brunet, Trinidad Caldés, Rosemarie Davidson, Jessica N. McAlpine, Jenny Lester, Niclas Håkansson, Kai-ren Ong, Ros Eeles, Francesmary Modugno, Martin Gore, Loic Le Marchand, Robert A. Vierkant, Wendy K. Chung, Christopher I. Amos, N. Charlotte Onland-Moret, Brita Arver, Marc Tischkowitz, Craig Luccarini, Daniel Barrowdale, Laima Tihomirova, Louise A. Brinton, Fergus J. Couch, Alfons Meindl, Nerea Larrañaga, Cristina Rodríguez-Antona, Alice S. Whittemore, Johanna I. Kiiski, Todd L. Edwards, Eric Hahnen, Grzegorz Sukiennicki, Els Van Nieuwenhuysen, Elizabeth J. van Rensburg, Michael Jones, Åke Borg, Edith Olah, Ute Hamann, Liv Cecilie Vestrheim Thomsen, Xiaoqing Chen, Ganna Chornokur, Minouk J. Schoemaker, Marc T. Goodman, Fanny Dao, Andrea Gehrig, Hagay Sobol, Nhu D. Le, Esther M. John, Adriaan Vanderstichele, Antonia Trichopoulou, Kunle Odunsi, Yukie Bean, David G. Huntsman, Lidia Pezzani, V. Wendy Setiawan, Marinus J. Blok, Yael Laitman, Mary Porteous, Patricia Harrington, Samantha Poblete, Shashikant Lele, Anne M. van Altena, Mingajeva Elvira, Lene Lundvall, Dominique Leroux, Annemarie H. van der Hout, Darya Prokofyeva, Debra Frost, Yoke-Eng Chiew, Gad Rennert, Stacey J. Winham, Muy-Kheng Tea, Kirsten B. Moysich, Angel Izquierdo, Anna H. Wu, Christine Rappaport-Fuerhauser, Peter Hillemanns, Rob B. van der Luijt, Gord Glendon, Jan Lubinński, Csilla Szabo, Gillian Mitchell, Andrea L. Richardson, Mark E. Robson, Roger L. Milne, Thomas Hansen, Francesca Damiola, Tameka Shelford, Natalia Antonenkova, Julie Lecarpentier, Paul A. James, Claudine Isaacs, Gianluca Severi, Maria A. Caligo, Teodora Goranova, Kate Lawrenson, Sandra Orsulic, Priyanka Sharma, Holly R. Harris, Peter A. Fasching, Christian Sutter, Torben A Kruse, Line Bjørge, Lesley McGuffog, Leon F.A.G. Massuger, Lynne R. Wilkens, Reidun K. Kopperud, Jillian Hung, Iain A. McNeish, Hanne Meijers-Heijboer, Uffe Birk Jensen, Diether Lambrechts, Johanna Rantala, Kelly-Anne Phillips, Michelle M.M. Woo, Kathryn L. Terry, Kathleen Claes, Ellen L. Goode, Olufunmilayo I. Olopade, Darcy L. Thull, Ailith Pirie, Sharon E. Johnatty, Soo Hwang Teo, Aimee A D'Aloisio, Evgeny N. Imyanitov, Domenico Palli, Andrew Berchuck, Banu Arun, Florentia Fostira, Jan Hauke, Jenny Chang-Claude, Pamela J. Thompson, Peter J. Hulick, Per Broberg, Arjen R. Mensenkamp, Xifeng Wu, Alicia A. Tone, Jacques Simard, Ursula Eilber, Jacek Gronwald, Kenneth Blankstein, James M. Flanagan, Alvaro N.A. Monteiro, Timea Pocza, Rita K. Schmutzler, Jeffrey N. Weitzel, Simon A. Gayther, William D. Foulkes, Valerie McGuire, Katherine L. Nathanson, Kevin H. Eng, Anna deFazio, Ian G. Campbell, Capucine Delnatte, Shan Wang-Gohrke, Jenna Lilyquist, Matti A. Rookus, Sakaeva Dina Damirovna, Laure Dossus, Simon G. Coetzee, Catherine J. Kennedy, Roberta B. Ness, James Paul, Andrew Lee, Linda E. Kelemen, Judith Balmaña, Anne-Marie Gerdes, Laure Barjhoux, Håkan Olsson, Lisa Walker, Doris Steinemann, Cecilia M. Dorfling, Julie M. Cunningham, Shelley S. Tworoger, Allan Jensen, Thomas A. Sellers, Javier Benitez, Anthony N. Karnezis, Hoda Anton-Culver, Carole Brewer, Siranoush Manoukian, Dorothea Wand, Ed Dicks, Antonis C. Antoniou, Amanda E. Toland, Anna Marie Mulligan, Päivi Kannisto, Rikki Cannioto, Bernd Dworniczak, Susan J. Ramus, Anna V. Tinker, Epidemiology and Data Science, Amsterdam Reproduction & Development (AR&D), CCA - Cancer biology and immunology, Amsterdam Neuroscience - Complex Trait Genetics, Human genetics, MUMC+: DA KG Lab Centraal Lab (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, Government of Canada, Cancer Research Foundation, Canadian Institutes of Health Research, National Institutes of Health (US), Fonds de Recherche du Québec, Ministry of Health and Welfare (South Korea), Associazione Italiana per la Ricerca sul Cancro, Deutsche Krebshilfe, Genome Canada, Medical Oncology, Clinical Genetics, Human Genetics, ARD - Amsterdam Reproduction and Development, Centre Léon Bérard [Lyon], Service de Génétique Oncologique, Institut Curie [Paris], CRLCC René Gauducheau, Centre International de Recherche contre le Cancer - International Agency for Research on Cancer (CIRC - IARC), Organisation Mondiale de la Santé / World Health Organization Office (OMS / WHO), Centre Hospitalier Métropole Savoie [Chambéry], CHU Grenoble, Cancer et génome: Bioinformatique, biostatistiques et épidémiologie d'un système complexe, Institut Curie [Paris]-MINES ParisTech - École nationale supérieure des mines de Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM), MINES ParisTech - École nationale supérieure des mines de Paris, Université Paris sciences et lettres (PSL), Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), CHU Saint-Etienne, Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Université Paris-Sud - Paris 11 - Faculté de médecine (UP11 UFR Médecine), Université Paris-Sud - Paris 11 (UP11), Institut Gustave Roussy (IGR), Université Paris-Saclay, Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), Tyrer, Jonathan [0000-0003-3724-4757], Dennis, Joe [0000-0003-4591-1214], Dicks, Ed [0000-0002-0617-0401], Lee, Andrew [0000-0003-0677-0252], Leslie, Goska [0000-0001-5756-6222], Brenton, James [0000-0002-5738-6683], Song, Honglin [0000-0001-5076-7371], Tischkowitz, Marc [0000-0002-7880-0628], Easton, Douglas [0000-0003-2444-3247], Antoniou, Antonis [0000-0001-9223-3116], Pharoah, Paul [0000-0001-8494-732X], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,endocrine system diseases ,Epidemiology ,Càncer d'ovari ,Genome-wide association study ,Carcinoma, Ovarian Epithelial ,Genome-wide association studies ,susceptibility ,Risk Factors ,Genotype ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Medicine and Health Sciences ,Neoplasms, Glandular and Epithelial ,POPULATION ,Genetics & Heredity ,Genetics ,Ovarian Neoplasms ,RISK ,education.field_of_study ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,cancer ,ovary ,BRCA1 Protein ,COMMON VARIANTS ,11 Medical And Health Sciences ,ASSOCIATION ,female genital diseases and pregnancy complications ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,3. Good health ,Serous fluid ,ovarian cancer ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Female ,Life Sciences & Biomedicine ,epithelial ovarian cancer, BRCA1, BRCA2 ,Population ,Telomere-Binding Proteins ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Meta-Analysis as Topic ,CLEAR-CELL CARCINOMA ,Ovarian cancer ,medicine ,Journal Article ,Humans ,BREAST-CANCER ,Genetic Predisposition to Disease ,Allele ,education ,Genotyping ,Alleles ,METAANALYSIS ,BRCA2 Protein ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Science & Technology ,MUTATIONS ,ENDOMETRIOSIS ,Biology and Life Sciences ,06 Biological Sciences ,medicine.disease ,030104 developmental biology ,Genetic Loci ,Mutation ,TELOMERE LENGTH ,Cancer research ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Developmental Biology ,Genome-Wide Association Study - Abstract
The OCAC OncoArray genotyping project: et al., To identify common alleles associated with different histotypes of epithelial ovarian cancer (EOC), we pooled data from multiple genome-wide genotyping projects totaling 25,509 EOC cases and 40,941 controls. We identified nine new susceptibility loci for different EOC histotypes: six for serous EOC histotypes (3q28, 4q32.3, 8q21.11, 10q24.33, 18q11.2 and 22q12.1), two for mucinous EOC (3q22.3 and 9q31.1) and one for endometrioid EOC (5q12.3). We then performed meta-analysis on the results for high-grade serous ovarian cancer with the results from analysis of 31,448 BRCA1 and BRCA2 mutation carriers, including 3,887 mutation carriers with EOC. This identified three additional susceptibility loci at 2q13, 8q24.1 and 12q24.31. Integrated analyses of genes and regulatory biofeatures at each locus predicted candidate susceptibility genes, including OBFC1, a new candidate susceptibility gene for low-grade and borderline serous EOC., The OCAC OncoArray genotyping project was funded through grants from the US National Institutes of Health (CA1X01HG007491-01 (C.I.A.), U19-CA148112 (T.A.S.), R01-CA149429 (C.M.P.) and R01-CA058598 (M.T.G.)); Canadian Institutes of Health Research (MOP-86727 (L.E.K.)); and the Ovarian Cancer Research Fund (A.B.). Funding for the CIMBA OncoArray genotyping was provided by the Government of Canada through Genome Canada and the Canadian Institutes of Health Research, the Ministère de l’Économie, de la Science et de l’Innovation du Québec through Génome Québec, the Quebec Breast Cancer Foundation for the PERSPECTIVE project, the US National Institutes of Health (CA1X01HG007491-01 (C.I.A.)), the Odense University Hospital Research Foundation (M.T.), the National R&D Program for Cancer Control, Ministry of Health and Welfare, Republic of Korea (1420190 (S.K.P.)), the Italian Association for Cancer Research (IG16933 (L.O.)) and German Cancer Aid (110837 (R.K.S.).
- Published
- 2017
- Full Text
- View/download PDF
34. Truncating mutations in the last exon ofNOTCH3cause lateral meningocele syndrome
- Author
-
Karen W. Gripp, Kimberly F. Doheny, Katia Sol-Church, Kristiina Avela, Nara Sobreira, Jacob Hogue, Daniela Alves, Katherine M. Robbins, Elaine H. Zackai, Deborah L. Stabley, Lynne M. Bird, P. Dane Witmer, and Outi Mäkitie
- Subjects
Genetics ,Pathology ,medicine.medical_specialty ,Hajdu–Cheney syndrome ,Point mutation ,Telecanthus ,Biology ,medicine.disease ,Hypotonia ,Lateral meningocele syndrome ,Skeletal disorder ,medicine ,Missense mutation ,Hypertelorism ,medicine.symptom ,Genetics (clinical) - Abstract
Lateral meningocele syndrome (LMS, OMIM%130720), also known as Lehman syndrome, is a very rare skeletal disorder with facial anomalies, hypotonia and meningocele-related neurologic dysfunction. The characteristic lateral meningoceles represent the severe end of the dural ectasia spectrum and are typically most severe in the lower spine. Facial features of LMS include hypertelorism and telecanthus, high arched eyebrows, ptosis, midfacial hypoplasia, micrognathia, high and narrow palate, low-set ears and a hypotonic appearance. Hyperextensibility, hernias and scoliosis reflect a connective tissue abnormality, and aortic dilation, a high-pitched nasal voice, wormian bones and osteolysis may be present. Lateral meningocele syndrome has phenotypic overlap with Hajdu–Cheney syndrome. We performed exome resequencing in five unrelated individuals with LMS and identified heterozygous truncating NOTCH3 mutations. In an additional unrelated individual Sanger sequencing revealed a deleterious variant in the same exon 33. In total, five novel de novo NOTCH3 mutations were identified in six unrelated patients. One had a 26 bp deletion (c.6461_6486del, p.G2154fsTer78), two carried the same single base pair insertion (c.6692_93insC, p.P2231fsTer11), and three individuals had a nonsense point mutation at c.6247A > T (pK2083*), c.6663C > G (p.Y2221*) or c.6732C > A, (p.Y2244*). All mutations cluster into the last coding exon, resulting in premature termination of the protein and truncation of the negative regulatory proline-glutamate-serine-threonine rich PEST domain. Our results suggest that mutant mRNA products escape nonsense mediated decay. The truncated NOTCH3 may cause gain-of-function through decreased clearance of the active intracellular product, resembling NOTCH2 mutations in the clinically related Hajdu–Cheney syndrome and contrasting the NOTCH3 missense mutations causing CADASIL. © 2014 Wiley Periodicals, Inc.
- Published
- 2014
- Full Text
- View/download PDF
35. Whole Exome Sequencing of Distant Relatives in Multiplex Families Implicates Rare Variants in Candidate Genes for Oral Clefts
- Author
-
Elisabeth Mangold, Markus M. Noethen, Joan E. Bailey-Wilson, Ingo Ruczinski, Alan F. Scott, Kimberly F. Doheny, Jacqueline B. Hetmanski, Qing Li, Margaret M. Parker, Cheryl D. Cropp, Alexandre Bureau, Jeffrey C. Murray, Silke Szymczak, Khalid Alqosayer, Terri H. Beaty, L. Leigh Field, Hasan Albacha-Hejazi, Mary L. Marazita, Yah Huei Wu-Chou, Hua Ling, Margaret A. Taub, and Kirsten U. Ludwig
- Subjects
Male ,Candidate gene ,Investigations ,Biology ,medicine.disease_cause ,symbols.namesake ,Antigens, CD ,Genetic linkage ,Ethnicity ,Genetics ,medicine ,Humans ,Exome ,Family ,Multiplex ,Gene ,Genetic Association Studies ,Exome sequencing ,Sanger sequencing ,Mutation ,Reproducibility of Results ,Sequence Analysis, DNA ,Cadherins ,Pedigree ,Cleft Palate ,symbols ,Female - Abstract
A dozen genes/regions have been confirmed as genetic risk factors for oral clefts in human association and linkage studies, and animal models argue even more genes may be involved. Genomic sequencing studies should identify specific causal variants and may reveal additional genes as influencing risk to oral clefts, which have a complex and heterogeneous etiology. We conducted a whole exome sequencing (WES) study to search for potentially causal variants using affected relatives drawn from multiplex cleft families. Two or three affected second, third, and higher degree relatives from 55 multiplex families were sequenced. We examined rare single nucleotide variants (SNVs) shared by affected relatives in 348 recognized candidate genes. Exact probabilities that affected relatives would share these rare variants were calculated, given pedigree structures, and corrected for the number of variants tested. Five novel and potentially damaging SNVs shared by affected distant relatives were found and confirmed by Sanger sequencing. One damaging SNV in CDH1, shared by three affected second cousins from a single family, attained statistical significance (P = 0.02 after correcting for multiple tests). Family-based designs such as the one used in this WES study offer important advantages for identifying genes likely to be causing complex and heterogeneous disorders.
- Published
- 2014
- Full Text
- View/download PDF
36. Design and Anticipated Outcomes of the eMERGE-PGx Project: A Multicenter Pilot for Preemptive Pharmacogenomics in Electronic Health Record Systems
- Author
-
Sarah C. Stallings, Eric B. Larson, Stuart A. Scott, John Connolly, Teri A. Manolio, Murray H. Brilliant, Ingrid A. Holm, Marylyn D. Ritchie, David Carrell, Dan M. Roden, Berta Almoguera, Laura J. Rasmussen-Torvik, Joshua C. Denny, Ana R. Mejia, Carlos J. Gallego, Wendy A. Wolf, James D. Ralston, Ariel Brautbar, Cassandra Perry, John B. Harley, Brendan J. Keating, Catherine A. McCarty, Rongling Li, Jonathan L. Haines, Josh F. Peterson, Jennifer A. Pacheco, Cynthia A. Prows, Kimberly F. Doheny, Erwin P. Bottinger, Rex L. Chisholm, Christopher G. Chute, Iftikhar J. Kullo, Jyotishman Pathak, Suzette J. Bielinski, Aida Vega, Melissa A. Basford, Luke V. Rasmussen, Shannon Manzi, Daniel Seung Kim, Omri Gottesman, Deborah A. Nickerson, David R. Crosslin, Steven E. Scherer, Adam S. Gordon, Senthilkumar Sadhasivam, Kathleen A. Leppig, Marc S. Williams, Simona Volpi, Maureen E. Smith, Simon Lin, Gail P. Jarvik, Vivian Pan, and Alexander A. Vinks
- Subjects
Male ,Gerontology ,Process management ,Adolescent ,Genotype ,Project implementation ,Process (engineering) ,Knowledge Bases ,Pilot Projects ,030226 pharmacology & pharmacy ,Clinical decision support system ,Article ,Young Adult ,03 medical and health sciences ,study design ,0302 clinical medicine ,Unknown Significance ,Drug Therapy ,Electronic health record ,Databases, Genetic ,Electronic Health Records ,Humans ,Medicine ,Pharmacology (medical) ,Sequence variation ,Child ,Genetic Association Studies ,Aged ,030304 developmental biology ,pharmacogenomics ,next generation sequencing ,Pharmacology ,0303 health sciences ,business.industry ,pre-emptive genotyping ,Genetic Variation ,Sequence Analysis, DNA ,Middle Aged ,Phenotype ,Pharmacogenetics ,Pharmacogenomics ,General partnership ,Female ,business - Abstract
We describe here the design and initial implementation of the eMERGE-PGx project. eMERGE-PGx, a partnership of the Electronic Medical Records and Genomics Network and the Pharmacogenomics Research Network, has three objectives: (i) to deploy PGRNseq, a next-generation sequencing platform assessing sequence variation in 84 proposed pharmacogenes, in nearly 9,000 patients likely to be prescribed drugs of interest in a 1- to 3-year time frame across several clinical sites; (ii) to integrate well-established clinically validated pharmacogenetic genotypes into the electronic health record with associated clinical decision support and to assess process and clinical outcomes of implementation; and (iii) to develop a repository of pharmacogenetic variants of unknown significance linked to a repository of electronic health record-based clinical phenotype data for ongoing pharmacogenomics discovery. We describe site-specific project implementation and anticipated products, including genetic variant and phenotype data repositories, novel variant association studies, clinical decision support modules, clinical and process outcomes, approaches to managing incidental findings, and patient and clinician education methods.
- Published
- 2014
- Full Text
- View/download PDF
37. Contents Vol. 5, 2014
- Author
-
Mathilde Huckert, Séraphin Nguefack, John F. Bateman, Renata Moldenhauer Minillo, Nara Sobreira, Vinciane Wouters, Corinne Stoetzel, Marie-Cécile Manière, Cedrik Tekendo-Ngongang, Virginie Laugel-Haushalter, Bee Chin Chen, Vincent Laugel, Kerry A. Miller, Rutger Meinsma, Kimberly F. Doheny, Cristina Has, Stefania Gimelli, Kurt N. Hetrick, Elisabeth Flori, Sophie Dahoun, Peter G. Farlie, David Valle, John B. Mulliken, Anne Dompmartin, Helen Mecili, Georgina Caruana, Martin Poot, Susan M. White, André B.P. van Kuilenburg, Laurence Myriam Boon, Decio Brunoni, Rowani Mohd Rawi, Agnès Bloch-Zupan, Ambroise Wonkam, Ilkka Kaitila, Raoul C.M. Hennekam, Druckerei Stückle, Judith Meijer, Megan F Welfare, Ana B. Alvarez Perez, Mustapha Amyere, Satz Mengensatzproduktion, Maria de Fátima de Faria Soares, John F. Bertram, Zornitza Stark, Odile Enjolras, Jean Muller, Ebtesam M. Abdalla, Miikka Vikkula, Tiong Yang Tan, Frédérique Sloan-Béna, Julie Jurgens, Catherine Godfraind, Pierre-Louis Docquier, Hua Ling, Ravi Savarirayan, Andrew A Heggie, Hélène Dollfus, and Trent Burgess
- Subjects
Genetics ,Genetics (clinical) - Published
- 2014
- Full Text
- View/download PDF
38. Novel Deletion of SERPINF1 Causes Autosomal Recessive Osteogenesis Imperfecta Type VI in Two Brazilian Families
- Author
-
Nara Sobreira, Ana B. Alvarez Perez, Kimberly F. Doheny, Decio Brunoni, Maria de Fátima de Faria Soares, Renata Moldenhauer Minillo, Kurt N. Hetrick, Hua Ling, Julie Jurgens, and David Valle
- Subjects
Genetics ,0303 health sciences ,business.industry ,030305 genetics & heredity ,medicine.disease ,Phenotype ,Bone morphogenetic protein 1 ,03 medical and health sciences ,PEDF ,PPIB ,Osteogenesis imperfecta ,Genotype ,Medicine ,business ,Gene ,Genetics (clinical) ,030304 developmental biology ,Founder effect - Abstract
Autosomal recessive osteogenesis imperfecta (OI) accounts for 10% of all OI cases, and, currently, mutations in 10 genes (CRTAP, LEPRE1, PPIB, SERPINH1, FKBP10, SERPINF1, SP7, BMP1, TMEM38B, and WNT1) are known to be responsible for this form of the disease. PEDF is a secreted glycoprotein of the serpin superfamily that maintains bone homeostasis and regulates osteoid mineralization, and it is encoded by SERPINF1, currently associated with OI type VI (MIM 172860). Here, we report a consanguineous Brazilian family in which multiple individuals from at least 4 generations are affected with a severe form of OI, and we also report an unrelated individual from the same small city in Brazil with a similar but more severe phenotype. In both families the same homozygous SERPINF1 19-bp deletion was identified which is not known in the literature yet. We described intra- and interfamilial clinical and radiological phenotypic variability of OI type VI caused by the same homozygous SERPINF1 19-bp deletion and suggest a founder effect. Furthermore, the SERPINF1 genotypes/phenotypes reported so far in the literature are reviewed.
- Published
- 2014
- Full Text
- View/download PDF
39. Imputation-Based Genomic Coverage Assessments of Current Human Genotyping Arrays
- Author
-
Elizabeth W. Pugh, Kimberly F. Doheny, Sarah C. Nelson, Cathy C. Laurie, Sharon R. Browning, Bruce S. Weir, Hua Ling, Jane Romm, and Cecelia A. Laurie
- Subjects
Linkage disequilibrium ,Genotyping Techniques ,Genome-wide association study ,Computational biology ,Investigations ,Biology ,Sensitivity and Specificity ,power ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,SNP microarrays ,Genetics ,Humans ,1000 Genomes Project ,Molecular Biology ,Allele frequency ,Genotyping ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,0303 health sciences ,genome-wide association study ,Genome, Human ,genomic coverage ,Minor allele frequency ,030217 neurology & neurosurgery ,Imputation (genetics) - Abstract
Microarray single-nucleotide polymorphism genotyping, combined with imputation of untyped variants, has been widely adopted as an efficient means to interrogate variation across the human genome. “Genomic coverage” is the total proportion of genomic variation captured by an array, either by direct observation or through an indirect means such as linkage disequilibrium or imputation. We have performed imputation-based genomic coverage assessments of eight current genotyping arrays that assay from ~0.3 to ~5 million variants. Coverage was determined separately in each of the four continental ancestry groups in the 1000 Genomes Project phase 1 release. We used the subset of 1000 Genomes variants present on each array to impute the remaining variants and assessed coverage based on correlation between imputed and observed allelic dosages. More than 75% of common variants (minor allele frequency > 0.05) are covered by all arrays in all groups except for African ancestry, and up to ~90% in all ancestries for the highest density arrays. In contrast, less than 40% of less common variants (0.01 < minor allele frequency < 0.05) are covered by low density arrays in all ancestries and 50–80% in high density arrays, depending on ancestry. We also calculated genome-wide power to detect variant-trait association in a case-control design, across varying sample sizes, effect sizes, and minor allele frequency ranges, and compare these array-based power estimates with a hypothetical array that would type all variants in 1000 Genomes. These imputation-based genomic coverage and power analyses are intended as a practical guide to researchers planning genetic studies.
- Published
- 2013
- Full Text
- View/download PDF
40. Generating Exome Enriched Sequencing Libraries from Formalin-Fixed, Paraffin-Embedded Tissue DNA for Next Generation Sequencing
- Author
-
Beth Marosy, Hua Ling, P. Dane Witmer, Brian Craig, Kurt N. Hetrick, Kimberly F. Doheny, Lawrence C. Brody, Elaine A. Ostrander, Janet L. Stanford, Sean Griffith, and Benjamin Myers
- Subjects
0301 basic medicine ,Cancer genome sequencing ,Tissue Fixation ,Biology ,DNA sequencing ,Article ,03 medical and health sciences ,Formaldehyde ,Exome Sequencing ,Genetics ,Humans ,Exome ,Genetics (clinical) ,Exome sequencing ,Illumina dye sequencing ,Gene Library ,Massive parallel sequencing ,Paraffin Embedding ,High-Throughput Nucleotide Sequencing ,DNA ,Molecular biology ,DNA sequencer ,030104 developmental biology ,Single cell sequencing ,Paraffin - Abstract
This unit describes a technique for generating exome-enriched sequencing libraries using DNA extracted from formalin-fixed paraffin-embedded (FFPE) samples. Utilizing commercially available kits, we present a low-input FFPE workflow starting with 50 ng of DNA. This procedure includes a repair step to address damage caused by FFPE preservation that improves sequence quality. Subsequently, libraries undergo an in-solution-targeted selection for exons, followed by sequencing using the Illumina next-generation short-read sequencing platform. © 2017 by John Wiley & Sons, Inc.
- Published
- 2017
41. Association analysis identifies 65 new breast cancer risk loci
- Author
-
Michael Jones, Dong-Young Noh, Martha J. Shrubsole, Chen-Yang Shen, Xiaoqing Chen, Esther M. John, Patricia Harrington, Quinten Waisfisz, Sue K. Park, Miriam Dwek, Christa Stegmaier, Sibylle Loibl, Wing-Yee Lo, Suleeporn Sangrajrang, Susan L. Neuhausen, Marina Bermisheva, Alicja Wolk, Christof Sohn, Keun-Young Yoo, Arja Jukkola-Vuorinen, Kathrin Thöne, James McKay, John J. Spinelli, Brian D. Carter, Elza Khusnutdinova, Francois Bacot, Paul L. Auer, Anna H. Wu, Christopher I. Amos, Johanna I. Kiiski, Hans Wildiers, Manjeet K. Bolla, Soo Hwang Teo, Julie M. Cunningham, Zhaoming Wang, Dieter Flesch-Janys, Argyrios Ziogas, Ann Smeets, Min Hyuk Lee, Xiaohong R. Yang, Carl Blomqvist, Natalia Antonenkova, Vassilios Georgoulias, Siranoush Manoukian, Anne Lise Børresen-Dale, Montserrat Garcia-Closas, Priyanka Sharma, Michael J. Kerin, Loic Le Marchand, Yoshio Kasuga, Michael Untch, Thomas Brüning, Natalia Bogdanova, Juliet D. French, Mark E. Sherman, Diana Eccles, Don M. Conroy, Marcia Adams, Pascal Guénel, Jonine D. Figueroa, Somchai Thanasitthichai, Jonathan Beesley, Paolo Peterlongo, Angela Cox, Graham G. Giles, Hans Christiansen, Irene L. Andrulis, Caroline Baynes, V. Shane Pankratz, Kristine Jones, Wei Zheng, Motoki Iwasaki, Rita K. Schmutzler, Julian Peto, Sara Margolin, Hedy S. Rennert, Hidemi Ito, Caroline Seynaeve, Jirong Long, Rachel Lloyd, Mark S. Goldberg, Javier Benitez, Børge G. Nordestgaard, Kathleen E. Malone, A. Heather Eliassen, Valerie Rhenius, Kristan J. Aronson, Maartje J. Hooning, Ursula Eilber, Christopher A. Haiman, Ji Yeob Choi, Jaana M. Hartikainen, Ian W. Brock, Barbara Burwinkel, Brigitte Rack, Mitul Shah, Matthias W. Beckmann, Belynda Hicks, Bernardo Bonanni, Alexander Hein, Leslie Bernstein, Christine L. Clarke, Emilie Cordina-Duverger, Sabine Behrens, Hoda Anton-Culver, Angela Brooks-Wilson, Bin Zhu, Gord Glendon, Banu Arun, José A. García-Sáenz, Jose Ignacio Arias Perez, Anne Grundy, Ans M.W. van den Ouweland, Nadege Presneau, Volker Arndt, Mikael Eriksson, Simon S. Cross, Craig Luccarini, Anthony J. Swerdlow, Veli-Matti Kosma, Hui Miao, Ming-Feng Hou, Hiroji Iwata, Christi J. van Asperen, Henrik Flyger, Jennifer Stone, Qin Wang, Penny Soucy, Heli Nevanlinna, Jane Heyworth, Shoichiro Tsugane, Annika Lindblom, Curtis Olswold, Trinidad Caldés, Laura Fachal, Ed Dicks, Shirley Hui, Katja Butterbach, Alison M. Dunning, Peter Devilee, Qiuyin Cai, Antonis C. Antoniou, Diether Lambrechts, Katarzyna Kaczmarek, Katri Pylkäs, Fredrick R. Schumacher, Arif B. Ekici, Aaron D. Norman, Jolanta Lissowska, Daniel C. Tessier, N Hamel, Paolo Radice, Håkan Olsson, Vessela N. Kristensen, Ling Tong, Harald Surowy, Rulla M. Tamimi, Jonathan Tyrer, Michael P. Lux, Stacey L. Edwards, Kathryn J. Ruddy, Giske Ursin, Myrto Barrdahl, Xiao-Ou Shu, Primitiva Menéndez, Sara Y. Brucker, Elad Ziv, Kenneth Muir, Eric Hahnen, Andy C. H. Lee, Rodney J. Scott, Lothar Haeberle, Darya Prokofyeva, Isabel dos-Santos-Silva, Peter Kraft, David G. Cox, Jong Won Lee, Sunil R. Lakhani, Kelly-Anne Phillips, Douglas F. Easton, Melissa C. Southey, Jan Lubinski, Jose E. Castelao, Hanne Meijers-Heijboer, Ivana Maleva Kostovska, Siddhartha Kar, Renske Keeman, Celine M. Vachon, Diana Torres, Stephen J. Chanock, Jack A. Taylor, Emiel J. Th. Rutgers, Chuen Neng Lee, Jason Vollenweider, Gadi Rennert, Jane Romm, Amy E. McCart Reed, Kee Seng Chia, Thomas Rüdiger, Anja Rudolph, Catriona McLean, David Van Den Berg, Christopher G. Scott, Mervi Grip, Lucy Xia, Georgia Chenevix-Trench, Robert Winqvist, Mary Beth Terry, Jenny Chang-Claude, Hans-Ulrich Ulmer, Mikael Hartman, Dona N. Ho, Maria Kabisch, Christoph Engel, Claire Mulot, Grethe I. Grenaker Alnæs, Arto Mannermaa, Nazneen Rahman, Tjoung-Won Park-Simon, Atocha Romero, Lin Fritschi, Manuela Gago-Dominguez, Matthias Ruebner, Valerie Gaborieau, Keitaro Matsuo, Asha Rostamianfar, Emmanouil Saloustros, Dijana Plaseska-Karanfilska, J.-P. Meyer, Sara Lindström, Annegien Broeks, Audrey Lemaçon, Tsun Leung Chan, Tongguang Cheng, Robert J. MacInnis, Habibul Ahsan, Federico Canzian, Paul Brennan, Mia M. Gaudet, David E. Goldgar, Anna Jakubowska, Paul D.P. Pharoah, Jacek Gronwald, Sung-Won Kim, Daehee Kang, Daniel O. Stram, Nichola Johnson, Rob B. van der Luijt, Sten Cornelissen, Camilla Wendt, Dylan M. Glubb, Olufunmilayo I. Olopade, Usha Menon, David J. Hunter, Kristiina Aittomäki, Jamie Allen, Chiu-Chen Tseng, Keith Humphreys, Edmond S. K. Ma, Peter Hillemanns, Hiltrud Brauch, Chia-Ni Hsiung, Sheila Seal, Junko Ishiguro, Dale P. Sandler, Peter Schürmann, Gary D. Bader, Sarah Stewart-Brown, Flavio Lejbkowicz, Marike Gabrielson, Wolfgang Janni, Judith S. Brand, Jingmei Li, Ava Kwong, Stig E. Bojesen, Ying Zheng, Mila Pinchev, Yu Tang Gao, Susan E. Hankinson, Elinor J. Sawyer, Per Broberg, Sofia Khan, Grace Sheng, Alfons Meindl, Margriet Collée, Jyh-Cherng Yu, Ute Krüger, Louise A. Brinton, Elizabeth W. Pugh, Thilo Dörk, Hilary K. Finucane, Peter A. Fasching, Shan Wang-Gohrke, Jenna Lilyquist, Muriel A. Adank, Marjanka K. Schmidt, Susan M. Gapstur, Sune F. Nielsen, Maya Ghoussaini, Minouk J. Schoemaker, Roger L. Milne, Jacques Simard, Clarice R. Weinberg, Kyriaki Michailidou, Shivaani Mariapun, Rob A. E. M. Tollenaar, Lizet E. van der Kolk, Nicola Miller, Anna González-Neira, Ian Tomlinson, Thérèse Truong, Ross L. Prentice, Anna Marie Mulligan, Jason S. Carroll, Taiki Yamaji, Carolina Ellberg, Mingajeva Elvira, Olivia Fletcher, Arnaud Droit, Per Hall, Maria Elena Martinez, Maria Tengström, Hui Cai, Xia Jiang, Janet E. Olson, Julia A. Knight, Nick Orr, Angel Carracedo, Guanmengqian Huang, Martine Dumont, Cheng Har Yip, Tom Maishman, Mary B. Daly, Artitaya Lophatananon, Niclas Håkansson, Steven N. Hart, Nur Aishah Taib, Andreas Schneeweiss, Daniel F. Schmidt, Daniel Vincent, Antoinette Hollestelle, Dimitrios Mavroudis, JoAnn E. Manson, Joe Dennis, Walter C. Willett, Rudolf Kaaks, Karen McCue, Enes Makalic, Kimberly F. Doheny, Eunjung Lee, Robert N. Hoover, Fergus J. Couch, Ute Hamann, Alice S. Whittemore, Hermann Brenner, Lorraine Durcan, Kamila Czene, Patrick Neven, John L. Hopper, Clinical Genetics, Medical Oncology, Human Genetics, CCA - Cancer biology and immunology, Dennis, Joe [0000-0003-4591-1214], Wang, Jean [0000-0002-9139-0627], Tyrer, Jonathan [0000-0003-3724-4757], Dicks, Ed [0000-0002-0617-0401], Lee, Andrew [0000-0003-0677-0252], Allen, Jamie [0000-0002-8677-2225], Fachal Vilar, Laura [0000-0002-7256-9752], Carroll, Jason [0000-0003-3643-0080], Rhenius, Valerie [0000-0003-4215-3235], Antoniou, Antonis [0000-0001-9223-3116], Pharoah, Paul [0000-0001-8494-732X], Dunning, Alison [0000-0001-6651-7166], Easton, Douglas [0000-0003-2444-3247], Apollo - University of Cambridge Repository, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, and Amsterdam Reproduction & Development (AR&D)
- Subjects
0301 basic medicine ,Multifactorial Inheritance ,Genome-wide association study ,Regulatory Sequences, Nucleic Acid ,skin and connective tissue diseases ,Cancer genetics ,Genetics ,Multidisciplinary ,medicine.diagnostic_test ,3. Good health ,Europe ,annotation ,Medical genetics ,Female ,Asian Continental Ancestry Group ,medicine.medical_specialty ,Asia ,architecture ,European Continental Ancestry Group ,ABCTB Investigators ,Locus (genetics) ,Breast Neoplasms ,Biology ,Polymorphism, Single Nucleotide ,Risk Assessment ,Article ,White People ,NBCS Collaborators ,03 medical and health sciences ,Breast cancer ,Asian People ,SDG 3 - Good Health and Well-being ,Genetic variation ,ConFab/AOCS Investigators ,expression ,medicine ,Genetic predisposition ,Journal Article ,Humans ,Computer Simulation ,Genetic Predisposition to Disease ,biological pathways ,Genetic association ,Genetic testing ,Genetic association study ,Binding Sites ,interaction networks ,medicine.disease ,comprehensive molecular portraits ,mutations ,susceptibility loci ,030104 developmental biology ,Genetic Loci ,genome-wide association ,protein ,Transcription Factors ,Genome-Wide Association Study - Abstract
Breast cancer risk is influenced by rare coding variants in susceptibility genes, such as BRCA1, and many common, mostly non-coding variants. However, much of the genetic contribution to breast cancer risk remains unknown. Here we report the results of a genome-wide association study of breast cancer in 122,977 cases and 105,974 controls of European ancestry and 14,068 cases and 13,104 controls of East Asian ancestry. We identified 65 new loci that are associated with overall breast cancer risk at P
- Published
- 2017
- Full Text
- View/download PDF
42. The OncoArray Consortium: a network for understanding the genetic architecture of common cancers
- Author
-
Katja Butterbach, Daniel C. Tessier, Dennis J. Hazelett, Stephen J. Chanock, Paul Brennan, Zhaoming Wang, Marc T. Goodman, Ali Amin Al Olama, Daniela Seminara, Tracy A. O'Mara, Craig Luccarini, Lesley McGuffog, David Van Den Berg, Andy C. H. Lee, James McKay, Stephen B. Gruber, Sue K. Park, Jack A. Taylor, Graham G. Giles, Michael F. Seldin, Alison M. Dunning, Zsofia Kote-Jarai, Sara Lindström, David E. Goldgar, Irene Brüske-Hohlfeld, Stig E. Bojesen, Paul D.P. Pharoah, Jonathan Marchini, Rosalind A. Eeles, Karoline Kuchenbaecker, Laura Fachal, Penny Soucy, Rayjean J. Hung, Ahsan Kamal, Marjorie J. Riggan, Xiangjun Xiao, Fergus J. Couch, Francois Bacot, Georgia Chenevix-Trench, John K. Field, Amanda B. Spurdle, Christopher A. Haiman, Julie M. Cunningham, Andrew Berchuck, Fredrick R. Schumacher, Elizabeth M. Gillanders, Simon A. Gayther, Belynda Hicks, Laura Ottini, David J. Hunter, Peter Kraft, Charlisse Caga-Anan, Jane Romm, Sylvie Laboissiere, Heike Bickeböller, Catherine M. Phelan, Jacques Simard, Graham Casey, Stephanie L. Schmit, Yafang Li, Sune F. Nielsen, Thomas A. Sellers, Hua Ling, Christopher K. Edlund, Yongyong Shi, Liesel M. FitzGerald, Stefanie A. Nelson, Rita K. Schmutzler, Gerhard A. Coetzee, Linda E. Kelemen, Kimberly F. Doheny, Elizabeth W. Pugh, Antonis C. Antoniou, Melanie Waldenberger, Judith L. Forman, Angela Risch, Ulrike Peters, Joe Dennis, David V. Conti, Daniel Vincent, Stephen Demetriades, Jinyoung Byun, Sara Benlloch, Tameka Shelford, Hongbing Shen, Douglas F. Easton, Mads Thomassen, Marcia Adams, Christopher I. Amos, Kenneth Offit, Kyriaki Michailidou, Judith Manz, Deborah J. Thompson, Dennis, Joe [0000-0003-4591-1214], Dunning, Alison [0000-0001-6651-7166], Fachal Vilar, Laura [0000-0002-7256-9752], Lee, Andrew [0000-0003-0677-0252], Thompson, Deborah [0000-0003-1465-5799], Amin Al Olama, Ali [0000-0002-7178-3431], Antoniou, Antonis [0000-0001-9223-3116], Pharoah, Paul [0000-0001-8494-732X], Easton, Douglas [0000-0003-2444-3247], and Apollo - University of Cambridge Repository
- Subjects
Male ,0301 basic medicine ,epistasis ,genetic epidemiology ,Genotype ,Epidemiology ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Risk Assessment ,Article ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Genetic Predisposition to Disease/epidemiology ,Genetic variation ,Mendelian randomization ,Prevalence ,Humans ,cancer ,Genetic Predisposition to Disease ,Selection, Genetic ,Genotyping ,Genetics ,ancestry ,Genetic Variation ,Polymorphism, Single Nucleotide/genetics ,Prognosis ,Genetic architecture ,3. Good health ,030104 developmental biology ,Genetic Variation/genetics ,Oncology ,030220 oncology & carcinogenesis ,OncoArray Consortium ,genetic architecture ,cancers ,Genome-Wide Association Study/methods ,Female ,DNA microarray ,Neoplasms/epidemiology ,Genome-Wide Association Study ,genetic susceptibility - Abstract
Background: Common cancers develop through a multistep process often including inherited susceptibility. Collaboration among multiple institutions, and funding from multiple sources, has allowed the development of an inexpensive genotyping microarray, the OncoArray. The array includes a genome-wide backbone, comprising 230,000 SNPs tagging most common genetic variants, together with dense mapping of known susceptibility regions, rare variants from sequencing experiments, pharmacogenetic markers and cancer related traits. Methods: The OncoArray can be genotyped using a novel technology developed by Illumina to facilitate efficient genotyping. The consortium developed standard approaches for selecting SNPs for study, for quality control of markers and for ancestry analysis. The array was genotyped at selected sites and with prespecified replicate samples to permit evaluation of genotyping accuracy among centers and by ethnic background. Results: The OncoArray consortium genotyped 447,705 samples. A total of 494,763 SNPs passed quality control steps with a sample success rate of 97% of the samples. Participating sites performed ancestry analysis using a common set of markers and a scoring algorithm based on principal components analysis. Conclusions: Results from these analyses will enable researchers to identify new susceptibility loci, perform fine mapping of new or known loci associated with either single or multiple cancers, assess the degree of overlap in cancer causation and pleiotropic effects of loci that have been identified for disease-specific risk, and jointly model genetic, environmental and lifestyle related exposures. Impact: Ongoing analyses will shed light on etiology and risk assessment for many types of cancer. TRICL (Transdisciplinary Research for Cancer of Lung) and International Lung Cancer Consortium (ILCCO): National Institute of Health U19 CA148127-01 (C.I. Amos, J. Byun, Y. Li, X. Xiao, J. L. Forman, A. Risch, H. Bickeböller, M. Waldenberger, I. Brüske, J. Manz, P. Brennan, R. Hung, H. Shen, Y. Shi, A. Kamal, C. I. Amos, J.K. Field), Canadian Cancer Society Research Institute (no. 020214, R. Hung). DRIVE (Discovery, Biology, and Risk of Inherited Variants in Breast Cancer): National Institute of Health U19 CA148065 (D.J. Hazelett, D. F. Easton, S. Lindström, P. Kraft, J. Dennis, A. Dunning, K. Michailidou, L. Fachal, S. Benlloch, J. Cunningham, K. Butterbach, K. Offit, R. Schmutzler, L. Ottini, D. Vincent). CORECT (ColoRectal Transdisciplinary Study): National Institute of Health U19 CA148107; R01 CA81488, P30 CA014089 (S. Gruber, U. Peters, G. Casey). ELLIPSE (ELLIPSE, Elucidating Loci in Prostate Cancer Susceptibility): This work was support by U19 CA148537(F. Schumacher, S.A. Gayther, S.L. Schmit, C.K. Edlund, D.J. Hazelett, G.A. Coetzee, C. Haiman, S. Demetriades, D. Van Den Berg). FOCI (Transdisciplinary Cancer Genetic Association and Interacting Studies): National Institutes of Health U19 CA148112- 01 (T.A. Sellers, C. Phelan), R01-CA122443(B. Hicks) ) P50-CA116201, CA192393 (F. Couch), P50-CA136393(J. Cunningham, F. Couch), OCAC - CA-149429 (C. Phelan). , P30-CA15083 (B. Hicks), Cancer Research UK (C490/A8339(A. Antoniou, P. Pharoah, A. Dunning), C490/A16561(P. Pharoah, A. Dunning), C490/A10119 (A. Dunning, P. Pharoah), C490/A10124 (A. Dunning, P. Pharoah)). ASTERISK: a Hospital Clinical Research Program (PHRC) and supported by the Regional Council of Pays de la Loire, the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (GEFLUC), the Association Anne de Bretagne Génétique and the Ligue Régionale Contre le Cancer (LRCC) (F. Bacot). DACHS: German Research Council (Deutsche Downloaded from cebp.aacrjournals.org on October 31, 2016. © 2016 American Association for Cancer Research. Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 3, 2016; DOI: 10.1158/1055-9965.EPI-16-0106 Forschungsgemeinschaft, BR 1704/6-1, BR 1704/6-3, BR 1704/6-4, and CH 117/1-1, C.K. Edlund), and the German Federal Ministry of Education and Research (01KH0404 and 01ER0814). Prostate Lung Colon Ovarian Screening trial: Intramural Research Program of the Division of Cancer Epidemiology and Genetics and supported by contracts from the Division of Cancer Prevention, National Cancer Institute, NIH, DHHS. The GC-HBOC (German Consortium of Hereditary Breast and Ovarian Cancer) is supported by the German Cancer Aid (grant no 110837, coordinator: R. Schmutzler, Cologne). The PERSPECTIVE (J. Simard) project was supported by the Government of Canada through Genome Canada and the Canadian Institutes of Health Research (MOP-86727, L.E. Kelemen), the Ministère de l’Économie, Innovation et Exportation du Québec through Genome Québec, and thee Quebec Breast Cancer Foundation, including additional funding from the Canadian Breast Cancer Foundation and the National Cancer Institute USA through Breast Cancer Family Registry Cohort (#1UM1CA164920-01A1). European Union Framework Programme for Research and Innovation (MSCA-IF-2014-EF-656144)(L. Fachal). The Breast Cancer Research Foundation, P30 CA08748, Andrew Sabin Foundation (K. Offit). CIDR genotyping for the Oncoarray was conducted under contract 268201200008I to K.Doheny and through grant 1X01HG007491- 01 to C.I. Amos. This is the author accepted manuscript. The final version is available from American Association for Cancer Research via http://dx.doi.org/10.1158/1055-9965.EPI-16-0106
- Published
- 2016
- Full Text
- View/download PDF
43. Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion
- Author
-
Anne U. Jackson, Jeroen R. Huyghe, Jane Romm, Johanna Kuusisto, Laura J. Scott, Lingyao Yang, Kimberly F. Doheny, Gonçalo R. Abecasis, Tanya M. Teslovich, Hyun Min Kang, Francis S. Collins, Mark J. Daly, Richard M. Watanabe, Michael Boehnke, Benjamin M. Neale, Marie P. Fogarty, Elizabeth W. Pugh, Hua Ling, Heather M. Stringham, Peter S. Chines, Markku Laakso, Alena Stančáková, Henna Cederberg, Martin L. Buchkovich, Karen L. Mohlke, Christian Fuchsberger, Roxann G. Ingersoll, Xueling Sim, and Ivy McMullen
- Subjects
Genetics ,education.field_of_study ,Insulin ,medicine.medical_treatment ,Population ,Biology ,medicine.disease ,medicine ,Metabolic syndrome ,education ,Exome ,Allele frequency ,Glycemic ,Insulin processing ,Proinsulin - Abstract
Karen Mohlke, Markku Laakso, Michael Boehnke and colleagues report the first application of the Illumina HumanExome Beadchip array, examining association with insulin and glycemic traits in 8,229 nondiabetic Finnish males from the population-based Metabolic Syndrome in Men (METSIM) study. They identify low-frequency coding variants at both known and newly associated loci with insulin processing and secretion.
- Published
- 2012
- Full Text
- View/download PDF
44. Detecting and Estimating Contamination of Human DNA Samples in Sequencing and Array-Based Genotype Data
- Author
-
Michael Boehnke, Kimberly F. Doheny, Kurt N. Hetrick, Goo Jun, Matthew Flickinger, Jane Romm, Gonçalo R. Abecasis, and Hyun Min Kang
- Subjects
Genetics ,Genotype ,Human dna ,Sequence analysis ,Sequence Analysis, DNA ,DNA Contamination ,Contamination ,Biology ,Article ,DNA sequencing ,Data sequences ,Diabetes Mellitus, Type 2 ,Humans ,Genetics(clinical) ,Sample contamination ,Genetics (clinical) - Abstract
DNA sample contamination is a serious problem in DNA sequencing studies and may result in systematic genotype misclassification and false positive associations. Although methods exist to detect and filter out cross-species contamination, few methods to detect within-species sample contamination are available. In this paper, we describe methods to identify within-species DNA sample contamination based on (1) a combination of sequencing reads and array-based genotype data, (2) sequence reads alone, and (3) array-based genotype data alone. Analysis of sequencing reads allows contamination detection after sequence data is generated but prior to variant calling; analysis of array-based genotype data allows contamination detection prior to generation of costly sequence data. Through a combination of analysis of in silico and experimentally contaminated samples, we show that our methods can reliably detect and estimate levels of contamination as low as 1%. We evaluate the impact of DNA contamination on genotype accuracy and propose effective strategies to screen for and prevent DNA contamination in sequencing studies.
- Published
- 2012
- Full Text
- View/download PDF
45. Interplay of Genetic Risk Factors (CHRNA5-CHRNA3-CHRNB4) and Cessation Treatments in Smoking Cessation Success
- Author
-
Megan E. Piper, Jen C. Wang, Dale S. Cannon, Kimberly F. Doheny, Li-Shiun Chen, Naomi Breslau, Eric O. Johnson, Nancy L. Saccone, Stephanie M. Gogarten, Robert B. Weiss, Laura J. Bierut, Alison Goate, and Timothy B. Baker
- Subjects
Bupropion ,medicine.medical_specialty ,biology ,business.industry ,CHRNA5 ,medicine.medical_treatment ,Article ,Psychiatry and Mental health ,Nicotinic agonist ,Internal medicine ,Genotype ,biology.protein ,medicine ,Physical therapy ,Smoking cessation ,Genetic risk ,business ,Early onset ,medicine.drug ,Nicotine replacement - Abstract
When smokers were separated by their nicotinic receptor gene variants, those with the low-risk genotype responded equally well to pharmacological treatments, including both nicotine replacement and bupropion, and nonpharmacological therapies. Those with the high-risk genotype, as identified by DNA sequencing, responded only to pharmacological treatments. Clinicians advising patients on smoking cessation can suspect genetic risk on the basis of early onset of heavy smoking and direct those smokers specifically to pharmacological treatments.
- Published
- 2012
- Full Text
- View/download PDF
46. CHRNB3 is more strongly associated with Fagerström Test for Cigarette Dependence-based nicotine dependence than cigarettes per day: phenotype definition changes genome-wide association studies results
- Author
-
Erin M. Ramos, Arpana Agrawal, Laura J. Bierut, Elizabeth W. Pugh, Kathleen K. Bucholz, William Howells, Samuel Kuperman, Rosalind J. Neuman, Siiri N. Bennett, Robert F. Krueger, Marc A. Schuckit, John P. Rice, Naomi Breslau, Teri A. Manolio, Howard J. Edenberg, Nancy L. Saccone, Cathy C. Laurie, Bernice Porjesz, Alison Goate, John I. Nurnberger, John Kramer, Kimberly F. Doheny, Scott F. Saccone, Victor Hesselbrock, Sarah M. Hartz, Laura Almasy, and Eric O. Johnson
- Subjects
business.industry ,Addiction ,media_common.quotation_subject ,Case-control study ,Medicine (miscellaneous) ,Genome-wide association study ,Odds ratio ,medicine.disease ,Phenotype ,Psychiatry and Mental health ,Meta-analysis ,Medicine ,business ,Nicotine dependence ,Genetic association ,media_common ,Demography - Abstract
Aims Nicotine dependence is a highly heritable disorder associated with severe medical morbidity and mortality. Recent meta-analyses have found novel genetic loci associated with cigarettes per day (CPD), a proxy for nicotine dependence. The aim of this paper is to evaluate the importance of phenotype definition (i.e. CPD versus Fagerstrom Test for Cigarette Dependence (FTCD) score as a measure of nicotine dependence) on genome-wide association studies of nicotine dependence. Design Genome-wide association study. Setting Community sample. Participants A total of 3365 subjects who had smoked at least one cigarette were selected from the Study of Addiction: Genetics and Environment (SAGE). Of the participants, 2267 were European Americans, 999 were African Americans. Measurements Nicotine dependence defined by FTCD score ≥4, CPD. Findings The genetic locus most strongly associated with nicotine dependence was rs1451240 on chromosome 8 in the region of CHRNB3 [odds ratio (OR) = 0.65, P = 2.4 × 10−8]. This association was further strengthened in a meta-analysis with a previously published data set (combined P = 6.7 × 10−16, total n = 4200). When CPD was used as an alternate phenotype, the association no longer reached genome-wide significance (β = −0.08, P = 0.0004). Conclusions Daily cigarette consumption and the Fagerstrom Test for Cigarette Dependence show different associations with polymorphisms in genetic loci.
- Published
- 2012
- Full Text
- View/download PDF
47. Detectable clonal mosaicism from birth to old age and its relationship to cancer
- Author
-
William J. Blot, Christopher I. Amos, Cynthia Regnier, Sara S. Strom, David R. Crosslin, Nadia N. Hansel, Janey L. Wiggs, Cathy C. Laurie, Andrew McDavid, Cecelia A. Laurie, Karl C. Desch, Eleanor Feingold, Kimberly F. Doheny, Lynn R. Goldin, Laura J. Bierut, Jun-Jun Li, Xiuwen Zheng, Hua Ling, Bruce S. Weir, Sarah M. Hartz, Lisa B. Signorello, Jeffrey C. Murray, Jeffrey E. Lee, Rasika A. Mathias, Louis R. Pasquale, Jenna Udren, Kristine R. Monroe, Ingo Ruczinski, Andrew Crenshaw, Bjarke Feenstra, Leila R. Zelnick, Maria Teresa Landi, Neil E. Caporaso, Teri A. Manolio, Kurt N. Hetrick, Terri H. Beaty, Kenneth Rice, Elizabeth W. Pugh, M. Geoffrey Hayes, Robert B. Scharpf, Sarah C. Nelson, Jae H. Kang, Sue A. Ingles, Venkatraman E. Seshan, Sonja I. Berndt, Matthew P. Conomos, Christopher A. Haiman, Li-E Wang, Gail P. Jarvik, Alan F. Scott, David Ginsburg, Loic Le Marchand, Stephen J. Chanock, Mads Melbye, Jess Shen, Brian E. Henderson, John A. Heit, Daniel B. Mirel, David M. Levine, Kathleen C. Barnes, Siiri N. Bennett, William L. Lowe, Qingyi Wei, Sebastian M. Armasu, Patrick J. Heagerty, Denise Daley, Nataliya Sharopova, Neal D. Freedman, Mariza de Andrade, Anastasia L. Wise, Mary L. Marazita, Stephanie M. Gogarten, and Caitlin P. McHugh
- Subjects
Adult ,Male ,Aging ,Adolescent ,DNA Copy Number Variations ,Physiology ,Genome-wide association study ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Genetics ,medicine ,Humans ,Child ,Gene ,Aged ,030304 developmental biology ,Genetic association ,Aged, 80 and over ,Chromosome Aberrations ,0303 health sciences ,Mosaicism ,Infant, Newborn ,Chromosome Mapping ,Infant ,Cancer ,Karyotype ,Middle Aged ,medicine.disease ,Confidence interval ,Uniparental disomy ,3. Good health ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Genome-Wide Association Study ,SNP array - Abstract
Clonal mosaicism for large chromosomal anomalies (duplications, deletions and uniparental disomy) was detected using SNP microarray data from over 50,000 subjects recruited for genome-wide association studies. This detection method requires a relatively high frequency of cells (>5–10%) with the same abnormal karyotype (presumably of clonal origin) in the presence of normal cells. The frequency of detectable clonal mosaicism in peripheral blood is low (
- Published
- 2012
- Full Text
- View/download PDF
48. Pitfalls of merging GWAS data: lessons learned in the eMERGE network and quality control procedures to maintain high data quality
- Author
-
Russell A. Wilke, Lana M. Olson, Hua Ling, Elizabeth W. Pugh, Teri A. Manolio, Yuki Bradford, Rongling Li, Loren L. Armstrong, Justin Paschall, Marylyn D. Ritchie, Lan Jiang, Christopher S. Carlson, Catherine A. McCarty, Gail P. Jarvik, Stephen D. Turner, Kimberly F. Doheny, Iftikhar J. Kullo, Andrew Crenshaw, Martha E. Matsumoto, M. Geoffrey Hayes, Daniel B. Mirel, Laura J. Rasmussen-Torvik, Andrew McDavid, Jonathan L. Haines, Rebecca L. Zuvich, Dana C. Crawford, Mariza de Andrade, Luke V. Rasmussen, and Suzette J. Bielinski
- Subjects
Epidemiology ,Quality control ,Genomics ,Genome-wide association study ,Biology ,computer.software_genre ,Pipeline (software) ,Biobank ,Data quality ,Human genome ,Data mining ,Genotyping ,computer ,Genetics (clinical) - Abstract
Genome-wide association studies (GWAS) are a useful approach in the study of the genetic components of complex phenotypes. Aside from large cohorts, GWAS have generally been limited to the study of one or a few diseases or traits. The emergence of biobanks linked to electronic medical records (EMRs) allows the efficient re-use of genetic data to yield meaningful genotype-phenotype associations for multiple phenotypes or traits. Phase I of the electronic MEdical Records and GEnomics (eMERGE-I) Network is a National Human Genome Research Institute (NHGRI)-supported consortium composed of five sites to perform various genetic association studies using DNA repositories and EMR systems. Each eMERGE site has developed EMR-based algorithms to comprise a core set of fourteen phenotypes for extraction of study samples from each site’s DNA repository. Each eMERGE site selected samples for a specific phenotype, and these samples were genotyped at either the Broad Institute or at the Center for Inherited Disease Research (CIDR) using the Illumina Infinium BeadChip technology. In all, approximately 17,000 samples from across the five sites were genotyped. A unified quality control (QC) pipeline was developed by the eMERGE Genomics Working Group and used to ensure thorough cleaning of the data. This process includes examination of sample quality, marker quality, and various batch effects. Upon completion of the genotyping and QC analyses for each site’s primary study, the eMERGE Coordinating Center merged the datasets from all five sites. This larger merged dataset re-entered the established eMERGE QC pipeline. Based on lessons learned during the process, additional analyses and QC checkpoints were added to the pipeline to ensure proper merging. Here we explore the challenges associated with combining datasets from different genotyping centers and describe the expansion to the eMERGE QC pipeline for merged datasets. These additional steps will be useful as the eMERGE project expands to include additional sites in eMERGE-II and also serve as a starting point for investigators merging multiple genotype data sets accessible through the National Center for Biotechnology Information (NCBI) in the database of Genotypes and Phenotypes (dbGaP). Our experience demonstrates that merging multiple datasets after additional QC can be an efficient use of genotype data despite new challenges that appear in the process.
- Published
- 2011
- Full Text
- View/download PDF
49. Genome-Wide Association Analysis of Ischemic Stroke in Young Adults
- Author
-
Christopher D. Anderson, Lynelle Cortellini, Braxton D. Mitchell, Nicole Dueker, Jeffrey R. O'Connell, Teri A. Manolio, O. Colin Stine, Mike A. Nalls, Sarah C. Nelson, Robert D. Brown, Karen L. Furie, Bradford B. Worrall, Hua Ling, Elizabeth W. Pugh, John W. Cole, Thomas G. Brott, Alessandro Biffi, Steven J. Kittner, Kimberly F. Doheny, Mary J. Sparks, Cathy C. Laurie, Stephen S. Rich, Yu-Ching Cheng, James F. Meschia, Patrick F. McArdle, Jess Shen, Jonathan Rosand, and Natalia S. Rost
- Subjects
Investigation ,Genetics ,0303 health sciences ,education.field_of_study ,Population ,FMNL2 ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Logistic regression ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Genotype ,SNP ,epidemiology ,brain infarction ,Young adult ,education ,Molecular Biology ,Genotyping ,030217 neurology & neurosurgery ,Genetics (clinical) ,030304 developmental biology - Abstract
Ischemic stroke (IS) is among the leading causes of death in Western countries. There is a significant genetic component to IS susceptibility, especially among young adults. To date, research to identify genetic loci predisposing to stroke has met only with limited success. We performed a genome-wide association (GWA) analysis of early-onset IS to identify potential stroke susceptibility loci. The GWA analysis was conducted by genotyping 1 million SNPs in a biracial population of 889 IS cases and 927 controls, ages 15–49 years. Genotypes were imputed using the HapMap3 reference panel to provide 1.4 million SNPs for analysis. Logistic regression models adjusting for age, recruitment stages, and population structure were used to determine the association of IS with individual SNPs. Although no single SNP reached genome-wide significance (P < 5 × 10−8), we identified two SNPs in chromosome 2q23.3, rs2304556 (in FMNL2; P = 1.2 × 10−7) and rs1986743 (in ARL6IP6; P = 2.7 × 10−7), strongly associated with early-onset stroke. These data suggest that a novel locus on human chromosome 2q23.3 may be associated with IS susceptibility among young adults.
- Published
- 2011
- Full Text
- View/download PDF
50. Genetic variants associated with the white blood cell count in 13,923 subjects in the eMERGE Network
- Author
-
Mariza de Andrade, David R. Crosslin, Sarah C. Nelson, Andrew Crenshaw, M. Geoffrey Hayes, Marylyn D. Ritchie, Eugene Hart, Xiuwen Zheng, Daniel B. Mirel, Iftikhar J. Kullo, Paul K. Crane, Catherine A. McCarty, Christopher S. Carlson, Gail P. Jarvik, Alexander Saip, Dana C. Crawford, Eric B. Larson, Noah Weston, Stephanie Pretel, Kimberly F. Doheny, Elizabeth W. Pugh, Katherine M. Newton, Rongling Li, Andrew McDavid, and Abel N. Kho
- Subjects
Adult ,Male ,Proteasome Endopeptidase Complex ,Genotype ,Black People ,Receptors, Cell Surface ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Medical Records ,White People ,Article ,Leukocyte Count ,Gene Frequency ,White blood cell ,Genetics ,medicine ,Humans ,Allele ,Alleles ,Genetics (clinical) ,Aged ,Genetic association ,Aged, 80 and over ,Principal Component Analysis ,Mediator Complex ,PSMD3 ,Genome, Human ,Genetic Variation ,Middle Aged ,Neoplasm Proteins ,medicine.anatomical_structure ,Immunology ,Absolute neutrophil count ,Female ,Duffy Blood-Group System ,Genome-Wide Association Study - Abstract
White blood cell count (WBC) is unique among identified inflammatory predictors of chronic disease in that it is routinely measured in asymptomatic patients in the course of routine patient care. We led a genome-wide association analysis to identify variants associated with WBC levels in 13,923 subjects in the electronic Medical Records and Genomics (eMERGE) Network. We identified two regions of interest that were each unique to subjects of genetically determined ancestry to the African continent (AA) or to the European continent (EA). WBC varies among different ancestry groups. Despite being ancestry specific, these regions were identifiable in the combined analysis. In AA subjects, the region surrounding the Duffy antigen/chemokine receptor gene (DARC) on 1q21 exhibited significant association (p value = 6.71e–55). These results validate the previously reported association between WBC and of the regulatory variant rs2814778 in the promoter region, which causes the Duffy negative phenotype (Fy−/−). A second missense variant (rs12075) is responsible for the two principal antigens, Fya and Fyb of the Duffy blood group system. The two variants, consisting of four alleles, act in concert to produce five antigens and subsequent phenotypes. We were able to identify the marginal and novel interaction effects of these two variants on WBC. In the EA subjects, we identified significantly associated SNPs tagging three separate genes in the 17q21 region: (1) GSDMA, (2) MED24, and (3) PSMD3. Variants in this region have been reported to be associated with WBC, neutrophil count, and inflammatory diseases including asthma and Crohn’s disease.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.