88 results on '"Michael L. Nickerson"'
Search Results
2. Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls
- Author
-
Jung Kim, Wen Luo, Mingyi Wang, Talia Wegman-Ostrosky, Megan N. Frone, Jennifer J. Johnston, Michael L. Nickerson, Melissa Rotunno, Shengchao A. Li, Maria I. Achatz, Seth A. Brodie, Michael Dean, Kelvin C. de Andrade, Fernanda P. Fortes, Matthew Gianferante, Payal Khincha, Mary L. McMaster, Lisa J. McReynolds, Alexander Pemov, Maisa Pinheiro, Karina M. Santiago, Blanche P. Alter, Neil E. Caporaso, Shahinaz M. Gadalla, Lynn R. Goldin, Mark H. Greene, Jennifer Loud, Xiaohong R. Yang, Neal D. Freedman, Susan M. Gapstur, Mia M. Gaudet, Donato Calista, Paola Ghiorzo, Maria Concetta Fargnoli, Eduardo Nagore, Ketty Peris, Susana Puig, Maria Teresa Landi, Belynda Hicks, Bin Zhu, Jia Liu, Joshua N. Sampson, Stephen J. Chanock, Lisa J. Mirabello, Lindsay M. Morton, Leslie G. Biesecker, Margaret A. Tucker, Sharon A. Savage, Alisa M. Goldstein, and Douglas R. Stewart
- Subjects
ACMG secondary findings ,Familial cancer exome ,Population study ,Variant classification ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Prior research has established that the prevalence of pathogenic/likely pathogenic (P/LP) variants across all of the American College of Medical Genetics (ACMG) Secondary Findings (SF) genes is approximately 0.8–5%. We investigated the prevalence of P/LP variants in the 24 ACMG SF v2.0 cancer genes in a family-based cancer research cohort (n = 1173) and in cancer-free ethnicity-matched controls (n = 982). Methods We used InterVar to classify variants and subsequently conducted a manual review to further examine variants of unknown significance (VUS). Results In the 24 genes on the ACMG SF v2.0 list associated with a cancer phenotype, we observed 8 P/LP unique variants (8 individuals; 0.8%) in controls and 11 P/LP unique variants (14 individuals; 1.2%) in cases, a non-significant difference. We reviewed 115 VUS. The median estimated per-variant review time required was 30 min; the first variant within a gene took significantly (p = 0.0009) longer to review (median = 60 min) compared with subsequent variants (median = 30 min). The concordance rate was 83.3% for the variants examined by two reviewers. Conclusion The 115 VUS required database and literature review, a time- and labor-intensive process hampered by the difficulty in interpreting conflicting P/LP determinations. By rigorously investigating the 24 ACMG SF v2.0 cancer genes, our work establishes a benchmark P/LP variant prevalence rate in a familial cancer cohort and controls.
- Published
- 2018
- Full Text
- View/download PDF
3. Diesel exhaust and bladder cancer risk by pathologic stage and grade subtypes
- Author
-
Stella Koutros, Manolis Kogevinas, Melissa C. Friesen, Patricia A. Stewart, Dalsu Baris, Margaret R. Karagas, Molly Schwenn, Alison Johnson, G.M. Monawar Hosain, Consol Serra, Adonina Tardon, Alfredo Carrato, Reina Garcia-Closas, Lee E. Moore, Michael L. Nickerson, Stephen M. Hewitt, Petra Lenz, Alan R. Schned, Josep Lloreta, Yves Allory, Haoyu Zhang, Nilanjan Chatterjee, Montserrat Garcia-Closas, Nathaniel Rothman, Núria Malats, and Debra T. Silverman
- Subjects
Environmental sciences ,GE1-350 - Abstract
Background: The International Agency for Research on Cancer (IARC) classifies diesel engine exhaust as carcinogenic to humans based on sufficient evidence for lung cancer. IARC noted, however, an increased risk of bladder cancer (based on limited evidence). Objective: To evaluate the association between quantitative, lifetime occupational diesel exhaust exposure and risk of urothelial cell carcinoma of the bladder (UBC) overall and according to pathological subtypes. Methods: Data from personal interviews with 1944 UBC cases, as well as formalin-fixed paraffin-embedded tumor tissue blocks, and 2135 controls were pooled from two case-control studies conducted in the U.S. and Spain. Lifetime occupational histories combined with exposure-oriented questions were used to estimate cumulative exposure to respirable elemental carbon (REC), a primary surrogate for diesel exhaust. Unconditional logistic regression and two-stage polytomous logistic regression were used to calculate odds ratios (ORs) and 95% confidence intervals (CIs), adjusting for smoking and other risk factors. Results: Exposure to cumulative REC was associated with an increased risk of UBC; workers with cumulative REC >396 μg/m3-years had an OR of 1.61 (95% CI, 1.08–2.40). At this level of cumulative exposure, similar results were observed in the U.S. and Spain, OR = 1.75 (95% CI, 0.97–3.15) and OR = 1.54 (95% CI, 0.89–2.68), respectively. In lagged analysis, we also observed a consistent increased risk among workers with cumulative REC >396 μg/m3-years (range of ORs = 1.52–1.93) for all lag intervals evaluated (5–40 years). When we accounted for tumor subtypes defined by stage and grade, a significant association between diesel exhaust exposure and UBC was apparent (global test for association p = 0.0019). Conclusions: Combining data from two large epidemiologic studies, our results provide further evidence that diesel exhaust exposure increases the risk of UBC. Keywords: Bladder cancer, Diesel exhaust, Occupation
- Published
- 2020
- Full Text
- View/download PDF
4. The Human TET2 Gene Contains Three Distinct Promoter Regions With Differing Tissue and Developmental Specificities
- Author
-
Hong Lou, Hongchuan Li, Kevin J. Ho, Luke L. Cai, Andy S. Huang, Tyler R. Shank, Michael R. Verneris, Michael L. Nickerson, Michael Dean, and Stephen K. Anderson
- Subjects
human ,TET2 ,alternative promoters ,demethylation ,differentiation ,mRNA isoforms ,Biology (General) ,QH301-705.5 - Abstract
Tet methylcytosine dioxygenase 2 (TET2) is a tumor suppressor gene that is inactivated in a wide range of hematological cancers. TET2 enzymatic activity converts 5-methylcytosine (5-mC) into 5-hydroxymethylcytosine (5-hmC), an essential step in DNA demethylation. Human TET2 is highly expressed in pluripotent cells and down-regulated in differentiated cells: however, transcriptional regulation of the human TET2 gene has not been investigated in detail. Here we define three promoters within a 2.5 kb region located ∼ 87 kb upstream of the first TET2 coding exon. The three promoters, designated as Pro1, Pro2, and Pro3, generate three alternative first exons, and their presence in TET2 mRNAs varies with cell type and developmental stage. In general, all three TET2 transcripts are more highly expressed in human tissues rich in hematopoietic stem cells, such as spleen and bone marrow, compared to other tissues, such as brain and kidney. Transcripts from Pro2 are expressed by a broad range of tissues and at a significantly higher level than Pro1 or Pro3 transcripts. Pro3 transcripts were highly expressed by embryoid bodies generated from the H9 ES cell line, and the major Pro3 transcript is an alternatively spliced mRNA isoform that produces a truncated TET2 protein lacking the catalytic domain. Our study demonstrates distinct tissue-specific mechanisms of TET2 transcriptional regulation during early pluripotent states and in differentiated cell types.
- Published
- 2019
- Full Text
- View/download PDF
5. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma
- Author
-
Christopher J. Ricketts, Aguirre A. De Cubas, Huihui Fan, Christof C. Smith, Martin Lang, Ed Reznik, Reanne Bowlby, Ewan A. Gibb, Rehan Akbani, Rameen Beroukhim, Donald P. Bottaro, Toni K. Choueiri, Richard A. Gibbs, Andrew K. Godwin, Scott Haake, A. Ari Hakimi, Elizabeth P. Henske, James J. Hsieh, Thai H. Ho, Rupa S. Kanchi, Bhavani Krishnan, David J. Kwaitkowski, Wembin Lui, Maria J. Merino, Gordon B. Mills, Jerome Myers, Michael L. Nickerson, Victor E. Reuter, Laura S. Schmidt, C. Simon Shelley, Hui Shen, Brian Shuch, Sabina Signoretti, Ramaprasad Srinivasan, Pheroze Tamboli, George Thomas, Benjamin G. Vincent, Cathy D. Vocke, David A. Wheeler, Lixing Yang, William T. Kim, A. Gordon Robertson, Paul T. Spellman, W. Kimryn Rathmell, W. Marston Linehan, Samantha J. Caesar-Johnson, John A. Demchok, Ina Felau, Melpomeni Kasapi, Martin L. Ferguson, Carolyn M. Hutter, Heidi J. Sofia, Roy Tarnuzzer, Zhining Wang, Liming Yang, Jean C. Zenklusen, Jiashan (Julia) Zhang, Sudha Chudamani, Jia Liu, Laxmi Lolla, Rashi Naresh, Todd Pihl, Qiang Sun, Yunhu Wan, Ye Wu, Juok Cho, Timothy DeFreitas, Scott Frazer, Nils Gehlenborg, Gad Getz, David I. Heiman, Jaegil Kim, Michael S. Lawrence, Pei Lin, Sam Meier, Michael S. Noble, Gordon Saksena, Doug Voet, Hailei Zhang, Brady Bernard, Nyasha Chambwe, Varsha Dhankani, Theo Knijnenburg, Roger Kramer, Kalle Leinonen, Yuexin Liu, Michael Miller, Sheila Reynolds, Ilya Shmulevich, Vesteinn Thorsson, Wei Zhang, Bradley M. Broom, Apurva M. Hegde, Zhenlin Ju, Anil Korkut, Jun Li, Han Liang, Shiyun Ling, Wenbin Liu, Yiling Lu, Kwok-Shing Ng, Arvind Rao, Michael Ryan, Jing Wang, John N. Weinstein, Jiexin Zhang, Adam Abeshouse, Joshua Armenia, Debyani Chakravarty, Walid K. Chatila, Ino de Bruijn, Jianjiong Gao, Benjamin E. Gross, Zachary J. Heins, Ritika Kundra, Konnor La, Marc Ladanyi, Augustin Luna, Moriah G. Nissan, Angelica Ochoa, Sarah M. Phillips, Francisco Sanchez-Vega, Chris Sander, Nikolaus Schultz, Robert Sheridan, S. Onur Sumer, Yichao Sun, Barry S. Taylor, Jioajiao Wang, Hongxin Zhang, Pavana Anur, Myron Peto, Paul Spellman, Christopher Benz, Joshua M. Stuart, Christopher K. Wong, Christina Yau, D. Neil Hayes, Joel S. Parker, Matthew D. Wilkerson, Adrian Ally, Miruna Balasundaram, Denise Brooks, Rebecca Carlsen, Eric Chuah, Noreen Dhalla, Robert Holt, Steven J.M. Jones, Katayoon Kasaian, Darlene Lee, Yussanne Ma, Marco A. Marra, Michael Mayo, Richard A. Moore, Andrew J. Mungall, Karen Mungall, Sara Sadeghi, Jacqueline E. Schein, Payal Sipahimalani, Angela Tam, Nina Thiessen, Kane Tse, Tina Wong, Ashton C. Berger, Andrew D. Cherniack, Carrie Cibulskis, Stacey B. Gabriel, Galen F. Gao, Gavin Ha, Matthew Meyerson, Steven E. Schumacher, Juliann Shih, Melanie H. Kucherlapati, Raju S. Kucherlapati, Stephen Baylin, Leslie Cope, Ludmila Danilova, Moiz S. Bootwalla, Phillip H. Lai, Dennis T. Maglinte, David J. Van Den Berg, Daniel J. Weisenberger, J. Todd Auman, Saianand Balu, Tom Bodenheimer, Cheng Fan, Katherine A. Hoadley, Alan P. Hoyle, Stuart R. Jefferys, Corbin D. Jones, Shaowu Meng, Piotr A. Mieczkowski, Lisle E. Mose, Amy H. Perou, Charles M. Perou, Jeffrey Roach, Yan Shi, Janae V. Simons, Tara Skelly, Matthew G. Soloway, Donghui Tan, Umadevi Veluvolu, Toshinori Hinoue, Peter W. Laird, Wanding Zhou, Michelle Bellair, Kyle Chang, Kyle Covington, Chad J. Creighton, Huyen Dinh, HarshaVardhan Doddapaneni, Lawrence A. Donehower, Jennifer Drummond, Robert Glenn, Walker Hale, Yi Han, Jianhong Hu, Viktoriya Korchina, Sandra Lee, Lora Lewis, Wei Li, Xiuping Liu, Margaret Morgan, Donna Morton, Donna Muzny, Jireh Santibanez, Margi Sheth, Eve Shinbrot, Linghua Wang, Min Wang, Liu Xi, Fengmei Zhao, Julian Hess, Elizabeth L. Appelbaum, Matthew Bailey, Matthew G. Cordes, Li Ding, Catrina C. Fronick, Lucinda A. Fulton, Robert S. Fulton, Cyriac Kandoth, Elaine R. Mardis, Michael D. McLellan, Christopher A. Miller, Heather K. Schmidt, Richard K. Wilson, Daniel Crain, Erin Curley, Johanna Gardner, Kevin Lau, David Mallery, Scott Morris, Joseph Paulauskis, Robert Penny, Candace Shelton, Troy Shelton, Mark Sherman, Eric Thompson, Peggy Yena, Jay Bowen, Julie M. Gastier-Foster, Mark Gerken, Kristen M. Leraas, Tara M. Lichtenberg, Nilsa C. Ramirez, Lisa Wise, Erik Zmuda, Niall Corcoran, Tony Costello, Christopher Hovens, Andre L. Carvalho, Ana C. de Carvalho, José H. Fregnani, Adhemar Longatto-Filho, Rui M. Reis, Cristovam Scapulatempo-Neto, Henrique C.S. Silveira, Daniel O. Vidal, Andrew Burnette, Jennifer Eschbacher, Beth Hermes, Ardene Noss, Rosy Singh, Matthew L. Anderson, Patricia D. Castro, Michael Ittmann, David Huntsman, Bernard Kohl, Xuan Le, Richard Thorp, Chris Andry, Elizabeth R. Duffy, Vladimir Lyadov, Oxana Paklina, Galiya Setdikova, Alexey Shabunin, Mikhail Tavobilov, Christopher McPherson, Ronald Warnick, Ross Berkowitz, Daniel Cramer, Colleen Feltmate, Neil Horowitz, Adam Kibel, Michael Muto, Chandrajit P. Raut, Andrei Malykh, Jill S. Barnholtz-Sloan, Wendi Barrett, Karen Devine, Jordonna Fulop, Quinn T. Ostrom, Kristen Shimmel, Yingli Wolinsky, Andrew E. Sloan, Agostino De Rose, Felice Giuliante, Marc Goodman, Beth Y. Karlan, Curt H. Hagedorn, John Eckman, Jodi Harr, Kelinda Tucker, Leigh Anne Zach, Brenda Deyarmin, Hai Hu, Leonid Kvecher, Caroline Larson, Richard J. Mural, Stella Somiari, Ales Vicha, Tomas Zelinka, Joseph Bennett, Mary Iacocca, Brenda Rabeno, Patricia Swanson, Mathieu Latour, Louis Lacombe, Bernard Têtu, Alain Bergeron, Mary McGraw, Susan M. Staugaitis, John Chabot, Hanina Hibshoosh, Antonia Sepulveda, Tao Su, Timothy Wang, Olga Potapova, Olga Voronina, Laurence Desjardins, Odette Mariani, Sergio Roman-Roman, Xavier Sastre, Marc-Henri Stern, Feixiong Cheng, Andrew Berchuck, Darell Bigner, Eric Lipp, Jeffrey Marks, Shannon McCall, Roger McLendon, Angeles Secord, Alexis Sharp, Madhusmita Behera, Daniel J. Brat, Amy Chen, Keith Delman, Seth Force, Fadlo Khuri, Kelly Magliocca, Shishir Maithel, Jeffrey J. Olson, Taofeek Owonikoko, Alan Pickens, Suresh Ramalingam, Dong M. Shin, Gabriel Sica, Erwin G. Van Meir, Hongzheng Zhang, Wil Eijckenboom, Ad Gillis, Esther Korpershoek, Leendert Looijenga, Wolter Oosterhuis, Hans Stoop, Kim E. van Kessel, Ellen C. Zwarthoff, Chiara Calatozzolo, Lucia Cuppini, Stefania Cuzzubbo, Francesco DiMeco, Gaetano Finocchiaro, Luca Mattei, Alessandro Perin, Bianca Pollo, Chu Chen, John Houck, Pawadee Lohavanichbutr, Arndt Hartmann, Christine Stoehr, Robert Stoehr, Helge Taubert, Sven Wach, Bernd Wullich, Witold Kycler, Dawid Murawa, Maciej Wiznerowicz, Ki Chung, W. Jeffrey Edenfield, Julie Martin, Eric Baudin, Glenn Bubley, Raphael Bueno, Assunta De Rienzo, William G. Richards, Steven Kalkanis, Tom Mikkelsen, Houtan Noushmehr, Lisa Scarpace, Nicolas Girard, Marta Aymerich, Elias Campo, Eva Giné, Armando López Guillermo, Nguyen Van Bang, Phan Thi Hanh, Bui Duc Phu, Yufang Tang, Howard Colman, Kimberley Evason, Peter R. Dottino, John A. Martignetti, Hani Gabra, Hartmut Juhl, Teniola Akeredolu, Serghei Stepa, Dave Hoon, Keunsoo Ahn, Koo Jeong Kang, Felix Beuschlein, Anne Breggia, Michael Birrer, Debra Bell, Mitesh Borad, Alan H. Bryce, Erik Castle, Vishal Chandan, John Cheville, John A. Copland, Michael Farnell, Thomas Flotte, Nasra Giama, Thai Ho, Michael Kendrick, Jean-Pierre Kocher, Karla Kopp, Catherine Moser, David Nagorney, Daniel O’Brien, Brian Patrick O’Neill, Tushar Patel, Gloria Petersen, Florencia Que, Michael Rivera, Lewis Roberts, Robert Smallridge, Thomas Smyrk, Melissa Stanton, R. Houston Thompson, Michael Torbenson, Ju Dong Yang, Lizhi Zhang, Fadi Brimo, Jaffer A. Ajani, Ana Maria Angulo Gonzalez, Carmen Behrens, Jolanta Bondaruk, Russell Broaddus, Bogdan Czerniak, Bita Esmaeli, Junya Fujimoto, Jeffrey Gershenwald, Charles Guo, Alexander J. Lazar, Christopher Logothetis, Funda Meric-Bernstam, Cesar Moran, Lois Ramondetta, David Rice, Anil Sood, Timothy Thompson, Patricia Troncoso, Anne Tsao, Ignacio Wistuba, Candace Carter, Lauren Haydu, Peter Hersey, Valerie Jakrot, Hojabr Kakavand, Richard Kefford, Kenneth Lee, Georgina Long, Graham Mann, Michael Quinn, Robyn Saw, Richard Scolyer, Kerwin Shannon, Andrew Spillane, onathan Stretch, Maria Synott, John Thompson, James Wilmott, Hikmat Al-Ahmadie, Timothy A. Chan, Ronald Ghossein, Anuradha Gopalan, Douglas A. Levine, Victor Reuter, Samuel Singer, Bhuvanesh Singh, Nguyen Viet Tien, Thomas Broudy, Cyrus Mirsaidi, Praveen Nair, Paul Drwiega, Judy Miller, Jennifer Smith, Howard Zaren, Joong-Won Park, Nguyen Phi Hung, Electron Kebebew, Adam R. Metwalli, Karel Pacak, Peter A. Pinto, Mark Schiffman, Nicolas Wentzensen, Robert Worrell, Hannah Yang, Marc Moncrieff, Chandra Goparaju, Jonathan Melamed, Harvey Pass, Natalia Botnariuc, Irina Caraman, Mircea Cernat, Inga Chemencedji, Adrian Clipca, Serghei Doruc, Ghenadie Gorincioi, Sergiu Mura, Maria Pirtac, Irina Stancul, Diana Tcaciuc, Monique Albert, Iakovina Alexopoulou, Angel Arnaout, John Bartlett, Jay Engel, Sebastien Gilbert, Jeremy Parfitt, Harman Sekhon, Doris M. Rassl, Robert C. Rintoul, Carlo Bifulco, Raina Tamakawa, Walter Urba, Nicholas Hayward, Henri Timmers, Anna Antenucci, Francesco Facciolo, Gianluca Grazi, Mirella Marino, Roberta Merola, Ronald de Krijger, Anne-Paule Gimenez-Roqueplo, Alain Piché, Simone Chevalier, Ginette McKercher, Kivanc Birsoy, Gene Barnett, Cathy Brewer, Carol Farver, Theresa Naska, Nathan A. Pennell, Daniel Raymond, Cathy Schilero, Kathy Smolenski, Felicia Williams, Carl Morrison, Jeffrey A. Borgia, Michael J. Liptay, Mark Pool, Christopher W. Seder, Kerstin Junker, Larsson Omberg, Mikhail Dinkin, George Manikhas, Domenico Alvaro, Maria Consiglia Bragazzi, Vincenzo Cardinale, Guido Carpino, Eugenio Gaudio, David Chesla, Sandra Cottingham, Michael Dubina, Fedor Moiseenko, Renumathy Dhanasekaran, Karl-Friedrich Becker, Klaus-Peter Janssen, Julia Slotta-Huspenina, Mohamed H. Abdel-Rahman, Dina Aziz, Sue Bell, Colleen M. Cebulla, Amy Davis, Rebecca Duell, J. Bradley Elder, Joe Hilty, Bahavna Kumar, James Lang, Norman L. Lehman, Randy Mandt, Phuong Nguyen, Robert Pilarski, Karan Rai, Lynn Schoenfield, Kelly Senecal, Paul Wakely, Paul Hansen, Ronald Lechan, James Powers, Arthur Tischler, William E. Grizzle, Katherine C. Sexton, Alison Kastl, Joel Henderson, Sima Porten, Jens Waldmann, Martin Fassnacht, Sylvia L. Asa, Dirk Schadendorf, Marta Couce, Markus Graefen, Hartwig Huland, Guido Sauter, Thorsten Schlomm, Ronald Simon, Pierre Tennstedt, Oluwole Olabode, Mark Nelson, Oliver Bathe, Peter R. Carroll, June M. Chan, Philip Disaia, Pat Glenn, Robin K. Kelley, Charles N. Landen, Joanna Phillips, Michael Prados, Jeffry Simko, Karen Smith-McCune, Scott VandenBerg, Kevin Roggin, Ashley Fehrenbach, Ady Kendler, Suzanne Sifri, Ruth Steele, Antonio Jimeno, Francis Carey, Ian Forgie, Massimo Mannelli, Michael Carney, Brenda Hernandez, Benito Campos, Christel Herold-Mende, Christin Jungk, Andreas Unterberg, Andreas von Deimling, Aaron Bossler, Joseph Galbraith, Laura Jacobus, Michael Knudson, Tina Knutson, Deqin Ma, Mohammed Milhem, Rita Sigmund, Rashna Madan, Howard G. Rosenthal, Clement Adebamowo, Sally N. Adebamowo, Alex Boussioutas, David Beer, Thomas Giordano, Anne-Marie Mes-Masson, Fred Saad, Therese Bocklage, Lisa Landrum, Robert Mannel, Kathleen Moore, Katherine Moxley, Russel Postier, Joan Walker, Rosemary Zuna, Michael Feldman, Federico Valdivieso, Rajiv Dhir, James Luketich, Edna M. Mora Pinero, Mario Quintero-Aguilo, Carlos Gilberto Carlotti, Jr., Jose Sebastião Dos Santos, Rafael Kemp, Ajith Sankarankuty, Daniela Tirapelli, James Catto, Kathy Agnew, Elizabeth Swisher, Jenette Creaney, Bruce Robinson, Carl Simon Shelley, Eryn M. Godwin, Sara Kendall, Cassaundra Shipman, Carol Bradford, Thomas Carey, Andrea Haddad, Jeffey Moyer, Lisa Peterson, Mark Prince, Laura Rozek, Gregory Wolf, Rayleen Bowman, Kwun M. Fong, Ian Yang, Robert Korst, J. Leigh Fantacone-Campbell, Jeffrey A. Hooke, Albert J. Kovatich, Craig D. Shriver, John DiPersio, Bettina Drake, Ramaswamy Govindan, Sharon Heath, Timothy Ley, Brian Van Tine, Peter Westervelt, Mark A. Rubin, Jung Il Lee, Natália D. Aredes, and Armaz Mariamidze
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Renal cell carcinoma (RCC) is not a single disease, but several histologically defined cancers with different genetic drivers, clinical courses, and therapeutic responses. The current study evaluated 843 RCC from the three major histologic subtypes, including 488 clear cell RCC, 274 papillary RCC, and 81 chromophobe RCC. Comprehensive genomic and phenotypic analysis of the RCC subtypes reveals distinctive features of each subtype that provide the foundation for the development of subtype-specific therapeutic and management strategies for patients affected with these cancers. Somatic alteration of BAP1, PBRM1, and PTEN and altered metabolic pathways correlated with subtype-specific decreased survival, while CDKN2A alteration, increased DNA hypermethylation, and increases in the immune-related Th2 gene expression signature correlated with decreased survival within all major histologic subtypes. CIMP-RCC demonstrated an increased immune signature, and a uniform and distinct metabolic expression pattern identified a subset of metabolically divergent (MD) ChRCC that associated with extremely poor survival. : Ricketts et al. find distinctive features of each RCC subtype, providing the foundation for development of subtype-specific therapeutic and management strategies. Somatic alteration of BAP1, PBRM1, and metabolic pathways correlates with subtype-specific decreased survival, while CDKN2A alteration, DNA hypermethylation, and Th2 immune signature correlate with decreased survival within all subtypes. Keywords: clear cell renal cell carcinoma, papillary renal cell carcinoma, chromophobe renal cell carcinoma, CDKN2A, DNA hypermethylation, immune signature, chromatin remodeling, TCGA, PanCanAtlas
- Published
- 2018
- Full Text
- View/download PDF
6. Supplementary Figure S1 from Concurrent Alterations in TERT, KDM6A, and the BRCA Pathway in Bladder Cancer
- Author
-
Dan Theodorescu, Michael Dean, M. Scott Lucia, Lee E. Moore, Zhiming Cai, Quan Zhou, Yingrui Li, Shirley X. Tsang, Guangwu Guo, James C. Costello, Charles Owens, Christina Ruiz-Rodriguez, Joseph Brown, Sevilay Turan, Michael G. Edwards, Kate M. Im, Garrett M. Dancik, and Michael L. Nickerson
- Abstract
The ubiquitin C (UBC) network in bladder cancer.
- Published
- 2023
7. Data from Concurrent Alterations in TERT, KDM6A, and the BRCA Pathway in Bladder Cancer
- Author
-
Dan Theodorescu, Michael Dean, M. Scott Lucia, Lee E. Moore, Zhiming Cai, Quan Zhou, Yingrui Li, Shirley X. Tsang, Guangwu Guo, James C. Costello, Charles Owens, Christina Ruiz-Rodriguez, Joseph Brown, Sevilay Turan, Michael G. Edwards, Kate M. Im, Garrett M. Dancik, and Michael L. Nickerson
- Abstract
Purpose: Genetic analysis of bladder cancer has revealed a number of frequently altered genes, including frequent alterations of the telomerase (TERT) gene promoter, although few altered genes have been functionally evaluated. Our objective is to characterize alterations observed by exome sequencing and sequencing of the TERT promoter, and to examine the functional relevance of histone lysine (K)–specific demethylase 6A (KDM6A/UTX), a frequently mutated histone demethylase, in bladder cancer.Experimental Design: We analyzed bladder cancer samples from 54 U.S. patients by exome and targeted sequencing and confirmed somatic variants using normal tissue from the same patient. We examined the biologic function of KDM6A using in vivo and in vitro assays.Results: We observed frequent somatic alterations in BRCA1 associated protein-1 (BAP1) in 15% of tumors, including deleterious alterations to the deubiquitinase active site and the nuclear localization signal. BAP1 mutations contribute to a high frequency of tumors with breast cancer (BRCA) DNA repair pathway alterations and were significantly associated with papillary histologic features in tumors. BAP1 and KDM6A mutations significantly co-occurred in tumors. Somatic variants altering the TERT promoter were found in 69% of tumors but were not correlated with alterations in other bladder cancer genes. We examined the function of KDM6A, altered in 24% of tumors, and show depletion in human bladder cancer cells, enhanced in vitro proliferation, in vivo tumor growth, and cell migration.Conclusions: This study is the first to identify frequent BAP1 and BRCA pathway alterations in bladder cancer, show TERT promoter alterations are independent of other bladder cancer gene alterations, and show KDM6A loss is a driver of the bladder cancer phenotype. Clin Cancer Res; 20(18); 4935–48. ©2014 AACR.
- Published
- 2023
8. Data from Targeted Deep Sequencing of Bladder Tumors Reveals Novel Associations between Cancer Gene Mutations and Mutational Signatures with Major Risk Factors
- Author
-
Michael Dean, Nathaniel Rothman, Debra T. Silverman, Ludmila Prokunina-Olsson, Montserrat Garcia-Closas, Kristine Jones, Alison Johnson, Molly Schwenn, Dalsu Baris, Larissa A. Pardo, Bin Zhu, Donghyuk Lee, Michael L. Nickerson, Lee E. Moore, Nina Rao, and Stella Koutros
- Abstract
Purpose:Exome- and whole-genome sequencing of muscle-invasive bladder cancer has revealed important insights into the molecular landscape; however, there are few studies of non–muscle-invasive bladder cancer with detailed risk factor information.Experimental Design:We examined the relationship between smoking and other bladder cancer risk factors and somatic mutations and mutational signatures in bladder tumors. Targeted sequencing of frequently mutated genes in bladder cancer was conducted in 322 formalin-fixed paraffin-embedded bladder tumors from a population-based case–control study. Logistic regression was used to calculate odds ratios (OR) and 95% confidence intervals (CI), evaluating mutations and risk factors. We used SignatureEstimation to extract four known single base substitution mutational signatures and Poisson regression to calculate risk ratios (RR) and 95% CIs, evaluating signatures and risk factors.Results:Non-silent KDM6A mutations were more common in females than males (OR = 1.83; 95% CI, 1.05–3.19). There was striking heterogeneity in the relationship between smoking status and established single base substitution signatures: current smoking status was associated with greater ERCC2-Signature mutations compared with former (P = 0.024) and never smoking (RR = 1.40; 95% CI, 1.09–1.80; P = 0.008), former smoking was associated with greater APOBEC-Signature13 mutations (P = 0.05), and never smoking was associated with greater APOBEC-Signature2 mutations (RR = 1.54; 95% CI, 1.17–2.01; P = 0.002). There was evidence that smoking duration (the component most strongly associated with bladder cancer risk) was associated with ERCC2-Signature mutations and APOBEC-Signature13 mutations among current (Ptrend = 0.005) and former smokers (P = 0.0004), respectively.Conclusions:These data quantify the contribution of bladder cancer risk factors to mutational burden and suggest different signature enrichments among never, former, and current smokers.
- Published
- 2023
9. Data from Improved Identification of von Hippel-Lindau Gene Alterations in Clear Cell Renal Tumors
- Author
-
Lee E. Moore, Frederic M. Waldman, Wong-Ho Chow, Nathaniel Rothman, Paul Brennan, Paolo Boffetta, Berton Zbar, Maria Merino, W. Marston Linehan, Gary F. Gerard, Rayjean Hung, Sara Karami, Jorge R. Toro, Laura S. Schmidt, Ivana Holcatova, Anush Mukeria, Dana Mates, Neonilia Szeszenia-Dabrowska, Marie Navratilova, Vladimir Bencko, Hellena Kollarova, Vladimir Janout, Vsevolod Matveev, David Zaridze, Sunil Mahurkar, Jeffrey A. Durocher, Yangu Shi, Erich Jaeger, and Michael L. Nickerson
- Abstract
Purpose: To provide a comprehensive, thorough analysis of somatic mutation and promoter hypermethylation of the von Hippel-Lindau (VHL) gene in the cancer genome, unique to clear cell renal cancer (ccRCC). Identify relationships between the prevalence of VHL gene alterations and alteration subtypes with patient and tumor characteristics.Experimental Design: As part of a large kidney cancer case-control study conducted in Central Europe, we analyzed VHL mutations and promoter methylation in 205 well-characterized, histologically confirmed patient tumor biopsies using a combination of sensitive, high-throughput methods (endonuclease scanning and Sanger sequencing) and analysis of 11 CpG sites in the VHL promoter.Results: We identified mutations in 82.4% of cases, the highest VHL gene mutation prevalence reported to date. Analysis of 11 VHL promoter CpG sites revealed that 8.3% of tumors were hypermethylated and all were mutation negative. In total, 91% of ccRCCs exhibited alteration of the gene through genetic or epigenetic mechanisms. Analysis of patient and tumor characteristics revealed that certain mutation subtypes were significantly associated with Fuhrman nuclear grade, metastasis, node positivity, and self-reported family history of RCC.Conclusion: Detection of VHL gene alterations using these accurate, sensitive, and practical methods provides evidence that the vast majority of histologically confirmed ccRCC tumors possess genetic or epigenetic alteration of the VHL gene and support the hypothesis that VHL alteration is an early event in ccRCC carcinogenesis. These findings also indicate that VHL molecular subtypes can provide a sensitive marker of tumor heterogeneity among histologically similar ccRCC cases for etiologic, prognostic, and translational studies.
- Published
- 2023
10. Driver genes exome sequencing reveals distinct variants in African Americans with colorectal neoplasia
- Author
-
Adeyinka O. Laiyemo, Hamed Azimi, Edward L. Lee, Hassan Brim, Hassan Ashktorab, Michael L. Nickerson, and Sudhir Varma
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,Population ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,targeted sequencing ,education ,neoplasms ,Exome sequencing ,African Americans ,education.field_of_study ,colon ,Cancer ,medicine.disease ,digestive system diseases ,3. Good health ,MSH6 ,030104 developmental biology ,druggable ,MSH3 ,030220 oncology & carcinogenesis ,KRAS ,actionable ,TCF7L2 ,Research Paper - Abstract
// Hassan Ashktorab 1 , Hamed Azimi 1 , Sudhir Varma 3 , Edward L. Lee 2 , Adeyinka O. Laiyemo 1 , Michael L. Nickerson 4 and Hassan Brim 2 1 Department of Medicine, Cancer Center, Howard University, Washington, DC, USA 2 Department of Pathology, Howard University College of Medicine, Washington, DC, USA 3 Hithru Analytics, LLC, Silver Spring, MD, USA 4 Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD, USA Correspondence to: Hassan Ashktorab, email: hashktorab@howard.edu Keywords: colon; targeted sequencing; African Americans; actionable; druggable Received: August 28, 2018 Accepted: January 31, 2019 Published: April 05, 2019 ABSTRACT Background: Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in the United States. African Americans are disproportionately affected by CRC. Our hypothesis is that driver genes with known and novel mutations have an impact on CRC outcome in this population. Therefore, we investigated the variants’ profiles in a panel of 15 CRC genes. Patients & Methods: Colorectal specimens (n=140) were analyzed by targeted exome sequencing using an Ion Torrent platform. Detected variants were validated in 36 samples by Illumina sequencing. The novel status of the validated variants was determined by comparison to publicly available databases. Annotated using ANNOVAR and in-silico functional analysis of these variants were performed to determine likely pathogenic variants. Results: Overall, 121 known and novel variants were validated: APC (27%), AMER1 (3%) , ARID1 (7%), MSH3 (12%), MSH6 (10%), BRAF (4%), KRAS (6%), FBXW7 (4%), PIK3CA (6%), SMAD4 (5%), SOX9 (2%), TCF7L2 (2%), TGFBR2 (5%), TP53 (7%). From these validated variants, 12% were novel in 8 genes (AMER1, APC, ARID1A, BRAF, MSH6, PIK3CA, SMAD4, and TCF7L2 ). Of the validated variants, 23% were non-synonymous, 14% were stopgains, 24% were synonymous and 39% were intronic variants. Conclusion: We here report the specifics of variants’ profiles of African Americans with colorectal lesions. Validated variants showed that Tumor Suppressor Genes (TSGs) APC and ARID1 and DNA Mismatch repair (MMR) genes MSH3 and MSH6 are the genes with the highest numbers of validated variants. Oncogenes KRAS and PIK3CA are also altered and likely participate in the increased proliferative potential of the mutated colonic epithelial cells in this population.
- Published
- 2019
11. Frequency of Pathogenic Germline Variants in Cancer-Susceptibility Genes in the Childhood Cancer Survivor Study
- Author
-
Jung Kim, Matthew Gianferante, Danielle M Karyadi, Stephen W Hartley, Megan N Frone, Wen Luo, Leslie L Robison, Gregory T Armstrong, Smita Bhatia, Michael Dean, Meredith Yeager, Bin Zhu, Lei Song, Joshua N Sampson, Yutaka Yasui, Wendy M Leisenring, Seth A Brodie, Kelvin C de Andrade, Fernanda P Fortes, Alisa M Goldstein, Payal P Khincha, Mitchell J Machiela, Mary L McMaster, Michael L Nickerson, Leatrisse Oba, Alexander Pemov, Maisa Pinheiro, Melissa Rotunno, Karina Santiago, Talia Wegman-Ostrosky, W Ryan Diver, Lauren Teras, Neal D Freedman, Belynda D Hicks, Mingyi Wang, Kristine Jones, Amy A Hutchinson, Casey Dagnall, Sharon A Savage, Margaret A Tucker, Stephen J Chanock, Lindsay M Morton, Douglas R Stewart, and Lisa Mirabello
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Genes, Recessive ,Penetrance ,Childhood Cancer Survivor Study ,Wilms Tumor ,Article ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Cancer Survivors ,CDKN2A ,Neoplasms ,Internal medicine ,Exome Sequencing ,Genetic predisposition ,medicine ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Child ,Germ-Line Mutation ,Exome sequencing ,Aged ,business.industry ,Lymphoma, Non-Hodgkin ,Cancer ,Sarcoma ,Wilms' tumor ,medicine.disease ,Pediatric cancer ,Kidney Neoplasms ,030104 developmental biology ,Case-Control Studies ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Background Pediatric cancers are the leading cause of death by disease in children despite improved survival rates overall. The contribution of germline genetic susceptibility to pediatric cancer survivors has not been extensively characterized. We assessed the frequency of pathogenic or likely pathogenic (P/LP) variants in 5451 long-term pediatric cancer survivors from the Childhood Cancer Survivor Study. Methods Exome sequencing was conducted on germline DNA from 5451 pediatric cancer survivors (cases who survived ≥5 years from diagnosis; n = 5105 European) and 597 European cancer-free adults (controls). Analyses focused on comparing the frequency of rare P/LP variants in 237 cancer-susceptibility genes and a subset of 60 autosomal dominant high-to-moderate penetrance genes, for both case-case and case-control comparisons. Results Of European cases, 4.1% harbored a P/LP variant in high-to-moderate penetrance autosomal dominant genes compared with 1.3% in controls (2-sided P = 3 × 10-4). The highest frequency of P/LP variants was in genes typically associated with adult onset rather than pediatric cancers, including BRCA1/2, FH, PALB2, PMS2, and CDKN2A. A statistically significant excess of P/LP variants, after correction for multiple tests, was detected in patients with central nervous system cancers (NF1, SUFU, TSC1, PTCH2), Wilms tumor (WT1, REST), non-Hodgkin lymphoma (PMS2), and soft tissue sarcomas (SDHB, DICER1, TP53, ERCC4, FGFR3) compared with other pediatric cancers. Conclusion In long-term pediatric cancer survivors, we identified P/LP variants in cancer-susceptibility genes not previously associated with pediatric cancer as well as confirmed known associations. Further characterization of variants in these genes in pediatric cancer will be important to provide optimal genetic counseling for patients and their families.
- Published
- 2021
12. Targeted Deep Sequencing of Bladder Tumors Reveals Novel Associations between Cancer Gene Mutations and Mutational Signatures with Major Risk Factors
- Author
-
Ludmila Prokunina-Olsson, Michael Dean, Lee E. Moore, Bin Zhu, Nina Rao, Alison Johnson, Michael L. Nickerson, DongHyuk Lee, Dalsu Baris, Nathaniel Rothman, Molly Schwenn, Montserrat Garcia-Closas, Kristine Jones, Stella Koutros, Larissa A. Pardo, and Debra T. Silverman
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Population ,Logistic regression ,Article ,symbols.namesake ,Risk Factors ,Internal medicine ,medicine ,Humans ,Poisson regression ,Risk factor ,education ,Exome ,Aged ,education.field_of_study ,Bladder cancer ,business.industry ,High-Throughput Nucleotide Sequencing ,Odds ratio ,Middle Aged ,medicine.disease ,Urinary Bladder Neoplasms ,Relative risk ,Case-Control Studies ,Mutation ,symbols ,Female ,business ,Genes, Neoplasm - Abstract
Purpose: Exome- and whole-genome sequencing of muscle-invasive bladder cancer has revealed important insights into the molecular landscape; however, there are few studies of non–muscle-invasive bladder cancer with detailed risk factor information. Experimental Design: We examined the relationship between smoking and other bladder cancer risk factors and somatic mutations and mutational signatures in bladder tumors. Targeted sequencing of frequently mutated genes in bladder cancer was conducted in 322 formalin-fixed paraffin-embedded bladder tumors from a population-based case–control study. Logistic regression was used to calculate odds ratios (OR) and 95% confidence intervals (CI), evaluating mutations and risk factors. We used SignatureEstimation to extract four known single base substitution mutational signatures and Poisson regression to calculate risk ratios (RR) and 95% CIs, evaluating signatures and risk factors. Results: Non-silent KDM6A mutations were more common in females than males (OR = 1.83; 95% CI, 1.05–3.19). There was striking heterogeneity in the relationship between smoking status and established single base substitution signatures: current smoking status was associated with greater ERCC2-Signature mutations compared with former (P = 0.024) and never smoking (RR = 1.40; 95% CI, 1.09–1.80; P = 0.008), former smoking was associated with greater APOBEC-Signature13 mutations (P = 0.05), and never smoking was associated with greater APOBEC-Signature2 mutations (RR = 1.54; 95% CI, 1.17–2.01; P = 0.002). There was evidence that smoking duration (the component most strongly associated with bladder cancer risk) was associated with ERCC2-Signature mutations and APOBEC-Signature13 mutations among current (Ptrend = 0.005) and former smokers (P = 0.0004), respectively. Conclusions: These data quantify the contribution of bladder cancer risk factors to mutational burden and suggest different signature enrichments among never, former, and current smokers.
- Published
- 2020
13. Differences in Tumor VHL Mutation and Hypoxia-inducible Factor 2α Expression Between African American and White Patients with Clear Cell Renal Cell Carcinoma
- Author
-
Mark P. Purdue, Lee J. Moore, Petra Lenz, Nathaniel Rothman, W. Marston Linehan, Catherine L. Callahan, and Michael L. Nickerson
- Subjects
African american ,Hypoxia-Inducible Factor 1 ,Mutation ,business.industry ,Urology ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,medicine.disease_cause ,Kidney Neoplasms ,White People ,Article ,Black or African American ,White (mutation) ,Clear cell renal cell carcinoma ,Hypoxia-inducible factors ,Von Hippel-Lindau Tumor Suppressor Protein ,VHL Mutation ,Basic Helix-Loop-Helix Transcription Factors ,Cancer research ,Carcinoma ,Humans ,Medicine ,business ,Carcinoma, Renal Cell - Published
- 2019
14. Distinctive DNA mismatch repair and APC rare variants in African Americans with colorectal neoplasia
- Author
-
Hassan Ashktorab, Hassan Brim, Payaam Tavakoli, Michael L. Nickerson, Sudhir Varma, and Hamed Azimi
- Subjects
African Americans ,0301 basic medicine ,Genetics ,colon ,Colorectal cancer ,targeted exome sequencing ,Cancer ,Biology ,medicine.disease ,3. Good health ,MSH6 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,MSH3 ,MSH2 ,030220 oncology & carcinogenesis ,medicine ,DNA mismatch repair ,Neoplastic transformation ,Exome sequencing ,Research Paper - Abstract
// Hassan Ashktorab 1 , Hamed Azimi 1 , Sudhir Varma 3 , Payaam Tavakoli 1 , Michael L. Nickerson 4 and Hassan Brim 2 1 Department of Medicine and Cancer Center, Washington, DC, USA 2 Department of Pathology, Howard University College of Medicine, Washington, DC, USA 3 Hithru LLC, Silver Spring, MD, USA 4 Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD, USA Correspondence to: Hassan Ashktorab, email: hashktorab@howard.edu Michael L. Nickerson, email: nickersonml@mail.nih.gov Keywords: targeted exome sequencing; colon; African Americans Received: January 25, 2017 Accepted: May 23, 2017 Published: October 07, 2017 ABSTRACT Purpose: African Americans have a higher incidence and mortality from colorectal cancer. This disparity might be due, in part, to the type of mutations in driver genes. In this study, we examined alterations specific to APC , MSH3 , and MSH6 genes using targeted exome sequencing to determine distinctive variants in the course of neoplastic transformation. Experimental Design: A total of 140 African American colon samples (30 normal, 21 adenomas, 33 advanced adenomas and 56 cancers) were used as our discovery set on an Ion Torrent platform. A 36 samples subset was resequenced on an Illumina platform for variants’ validation. Bioinformatics analyses were performed and novel validated variants are reported. Results: Two novel MSH6 variants were validated and mapped to the MutS-V region near the MSH2 binding site. For MSH3, 4 known variants were validated and were located in exon 10 (3 non-synonymous) and exon 18 (1 synonymous). As for APC , 20 variants were validated with 4 novel variants: 3 stopgain and 1 non-synonymous. These variants mapped prior to and on the Armadillo repeats region, to the 15-amino acid repeat region, and to the 20-amino acid repeats region, respectively. Conclusion: We defined novel variants that target DNA mismatch repair and APC genes in African Americans with colorectal lesions. A greater frequency of variants in genes encoding DNA mismatch repair functions and APC likely plays major roles in colorectal cancer initiation and higher incidence of the disease in African Americans.
- Published
- 2017
15. The Human
- Author
-
Hong, Lou, Hongchuan, Li, Kevin J, Ho, Luke L, Cai, Andy S, Huang, Tyler R, Shank, Michael R, Verneris, Michael L, Nickerson, Michael, Dean, and Stephen K, Anderson
- Subjects
Cell and Developmental Biology ,TET2 ,demethylation ,human ,differentiation ,alternative promoters ,mRNA isoforms ,Original Research - Abstract
Tet methylcytosine dioxygenase 2 (TET2) is a tumor suppressor gene that is inactivated in a wide range of hematological cancers. TET2 enzymatic activity converts 5-methylcytosine (5-mC) into 5-hydroxymethylcytosine (5-hmC), an essential step in DNA demethylation. Human TET2 is highly expressed in pluripotent cells and down-regulated in differentiated cells: however, transcriptional regulation of the human TET2 gene has not been investigated in detail. Here we define three promoters within a 2.5 kb region located ∼ 87 kb upstream of the first TET2 coding exon. The three promoters, designated as Pro1, Pro2, and Pro3, generate three alternative first exons, and their presence in TET2 mRNAs varies with cell type and developmental stage. In general, all three TET2 transcripts are more highly expressed in human tissues rich in hematopoietic stem cells, such as spleen and bone marrow, compared to other tissues, such as brain and kidney. Transcripts from Pro2 are expressed by a broad range of tissues and at a significantly higher level than Pro1 or Pro3 transcripts. Pro3 transcripts were highly expressed by embryoid bodies generated from the H9 ES cell line, and the major Pro3 transcript is an alternatively spliced mRNA isoform that produces a truncated TET2 protein lacking the catalytic domain. Our study demonstrates distinct tissue-specific mechanisms of TET2 transcriptional regulation during early pluripotent states and in differentiated cell types.
- Published
- 2019
16. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide
- Author
-
Carla Daniela Robles-Espinoza, Hilary Racher, Luca Mastracci, Robert Pilarski, Frederick H. Davidorf, Saleem Taibjee, William Glasson, Martine J. Jager, Jo Burke, Ivana K. Kim, Lisa Golmard, Rajmohan Murali, Hayley Hamilton, Giulia Ciccarese, Jane M. Palmer, Madeleine Howlie, Arnaud de la Fouchardière, David J. Adams, Tero Kivelä, Sonika Dahiya, Peter Johansson, Lorenza Pastorino, Sebastian Walpole, Annelies de Klein, Bruna Dalmasso, Anne Marie Lane, Marina Marinkovic, William Bruno, Judith Symmons, Sonja Klebe, Julia Newton-Bishop, Marc-Henri Stern, Michael R. Speicher, Joni A. Turunen, Colleen M. Cebulla, Dominique Stoppa-Lyonnet, Carsten Bergmann, Hildur Helgadottir, Mohamed H. Abdel-Rahman, S.J. O’Shea, Meredith Stautberg, Ching-Ni Njauw, Jens Folke Kiilgaard, Pauliina Repo, Erin M. Garfield, Paola Queirolo, Rana'a T. Al-Jamal, Remco van Doorn, Antonia L. Pritchard, Gregorius P M Luyten, Michael L. Nickerson, Mitchell Cheung, Pedram Gerami, Miriam Potrony, Paola Ghiorzo, Odile Cabaret, Julian Adlard, Veronica Höiom, Hensin Tsao, Cindy Chau, J. William Harbour, Irma Dianzani, Joseph R. Testa, Brigitte Bressac-de Paillerets, Sunil K Warrier, Maartje Nielsen, Marta Betti, Susana Puig, Emine Kilic, Sandro Santagata, Karin Wadt, Nicholas K. Hayward, Virginia Andreotti, Raul Ossio, Natasha M. van Poppelen, Nicola K. Poplawski, Reetta Stiina Järvinen, Clinical Genetics, and Ophthalmology
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Genotype ,Reviews ,Biology ,Risk Assessment ,Germline ,Age of Onset ,Alleles ,Female ,Gene Frequency ,Humans ,Neoplastic Syndromes, Hereditary ,Phenotype ,Tumor Suppressor Proteins ,Ubiquitin Thiolesterase ,Genetic Association Studies ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Genotype-phenotype distinction ,Germline mutation ,Missense mutation ,Neoplastic Syndromes ,Allele ,BAP1 ,3. Good health ,Hereditary ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer research - Abstract
BACKGROUND: The BRCA1-associated protein-1 (BAP1) tumor predisposition syndrome (BAP1-TPDS) is a hereditary tumor syndrome caused by germline pathogenic variants in BAP1 encoding a tumor suppressor associated with uveal melanoma, mesothelioma, cutaneous melanoma, renal cell carcinoma, and cutaneous BAP1-inactivated melanocytic tumors. However, the full spectrum of tumors associated with the syndrome is yet to be determined. Improved understanding of the BAP1-TPDS is crucial for appropriate clinical management of BAP1 germline variant carriers and their families, including genetic counseling and surveillance for new tumors. METHODS: We collated germline variant status, tumor diagnoses, and information on BAP1 immunohistochemistry or loss of somatic heterozygosity on 106 published and 75 unpublished BAP1 germline variant-positive families worldwide to better characterize the genotypes and phenotypes associated with the BAP1-TPDS. Tumor spectrum and ages of onset were compared between missense and null variants. All statistical tests were two-sided. RESULTS: The 181 families carried 140 unique BAP1 germline variants. The collated data confirmed the core tumor spectrum associated with the BAP1-TPDS and showed that some families carrying missense variants can exhibit this phenotype. A variety of noncore BAP1-TPDS -associated tumors were found in families of variant carriers. Median ages of onset of core tumor types were lower in null than missense variant carriers for all tumors combined (P
- Published
- 2018
17. Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls
- Author
-
Ketty Peris, Jennifer T. Loud, Maisa Pinheiro, Alexander Pemov, Stephen J. Chanock, Seth A. Brodie, Susana Puig, Jennifer J. Johnston, Maria Concetta Fargnoli, Joshua N. Sampson, Xiaohong R. Yang, Payal P. Khincha, Lindsay M. Morton, Sharon A. Savage, Talia Wegman-Ostrosky, Megan N. Frone, Neil E. Caporaso, Eduardo Nagore, Lisa J. McReynolds, Shahinaz M. Gadalla, Mingyi Wang, Michael L. Nickerson, Paola Ghiorzo, Mark H. Greene, Mary L. McMaster, Kelvin C. de Andrade, Mia M. Gaudet, Michael Dean, Bin Zhu, Leslie G. Biesecker, Donato Calista, Belynda Hicks, Margaret A. Tucker, Jia Liu, Wen Luo, Maria Isabel Achatz, Matthew Gianferante, Lisa Mirabello, Neal D. Freedman, Melissa Rotunno, Lynn R. Goldin, Jung Kim, Alisa M. Goldstein, Shengchao A. Li, Blanche P. Alter, Fernanda P. Fortes, Douglas R. Stewart, Maria Teresa Landi, Susan M. Gapstur, and Karina Miranda Santiago
- Subjects
0301 basic medicine ,Oncology ,Male ,DNA Mutational Analysis ,Prevalence ,lcsh:Medicine ,Cohort Studies ,Familial cancer exome ,Neoplasms ,Ethnicity ,Genetics (clinical) ,Single Nucleotide ,3. Good health ,Variant classification ,Cohort ,Molecular Medicine ,Medical genetics ,Population study ,Female ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,medicine.medical_specialty ,lcsh:QH426-470 ,Concordance ,Ethnic Groups ,ACMG secondary findings ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Aged ,Genes, Neoplasm ,Humans ,Genetic Predisposition to Disease ,Mutation ,Internal medicine ,Genetics ,medicine ,Polymorphism ,Molecular Biology ,Gene ,business.industry ,Research ,lcsh:R ,Cancer ,medicine.disease ,Human genetics ,lcsh:Genetics ,030104 developmental biology ,Genes ,ACMG secondary findings, Familial cancer exome, Population study, Variant classification ,Neoplasm ,business - Abstract
Background Prior research has established that the prevalence of pathogenic/likely pathogenic (P/LP) variants across all of the American College of Medical Genetics (ACMG) Secondary Findings (SF) genes is approximately 0.8–5%. We investigated the prevalence of P/LP variants in the 24 ACMG SF v2.0 cancer genes in a family-based cancer research cohort (n = 1173) and in cancer-free ethnicity-matched controls (n = 982). Methods We used InterVar to classify variants and subsequently conducted a manual review to further examine variants of unknown significance (VUS). Results In the 24 genes on the ACMG SF v2.0 list associated with a cancer phenotype, we observed 8 P/LP unique variants (8 individuals; 0.8%) in controls and 11 P/LP unique variants (14 individuals; 1.2%) in cases, a non-significant difference. We reviewed 115 VUS. The median estimated per-variant review time required was 30 min; the first variant within a gene took significantly (p = 0.0009) longer to review (median = 60 min) compared with subsequent variants (median = 30 min). The concordance rate was 83.3% for the variants examined by two reviewers. Conclusion The 115 VUS required database and literature review, a time- and labor-intensive process hampered by the difficulty in interpreting conflicting P/LP determinations. By rigorously investigating the 24 ACMG SF v2.0 cancer genes, our work establishes a benchmark P/LP variant prevalence rate in a familial cancer cohort and controls. Electronic supplementary material The online version of this article (10.1186/s13073-018-0607-5) contains supplementary material, which is available to authorized users.
- Published
- 2018
18. Molecular analysis of urothelial cancer cell lines for modeling tumor biology and drug response
- Author
-
Michael Dean, Z Cai, Michael L. Nickerson, James C. Costello, S X Tsang, Sevilay Turan, K M Im, S Wu, Charles Owens, N Witte, Dan Theodorescu, and K Misner
- Subjects
0301 basic medicine ,Urologic Neoplasms ,Cancer Research ,DNA Copy Number Variations ,Somatic cell ,DNA repair ,Antineoplastic Agents ,Biology ,Models, Biological ,Polymorphism, Single Nucleotide ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,CDKN2A ,Cell Line, Tumor ,CDKN2B ,Genetics ,medicine ,Cluster Analysis ,Humans ,Exome ,skin and connective tissue diseases ,Molecular Biology ,Gene ,TOR Serine-Threonine Kinases ,Chromosome Mapping ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Genomics ,DNA Methylation ,Cell cycle ,Prognosis ,medicine.disease ,Primary tumor ,Molecular biology ,3. Good health ,Treatment Outcome ,030104 developmental biology ,Mutation ,DNA methylation ,Original Article ,Chromosome Deletion ,Chromosomes, Human, Pair 9 ,Biomarkers ,Signal Transduction - Abstract
The utility of tumor-derived cell lines is dependent on their ability to recapitulate underlying genomic aberrations and primary tumor biology. Here, we sequenced the exomes of 25 bladder cancer (BCa) cell lines and compared mutations, copy number alterations (CNAs), gene expression and drug response to BCa patient profiles in The Cancer Genome Atlas (TCGA). We observed a mutation pattern associated with altered CpGs and APOBEC-family cytosine deaminases similar to mutation signatures derived from somatic alterations in muscle-invasive (MI) primary tumors, highlighting a major mechanism(s) contributing to cancer-associated alterations in the BCa cell line exomes. Non-silent sequence alterations were confirmed in 76 cancer-associated genes, including mutations that likely activate oncogenes TERT and PIK3CA, and alter chromatin-associated proteins (MLL3, ARID1A, CHD6 and KDM6A) and established BCa genes (TP53, RB1, CDKN2A and TSC1). We identified alterations in signaling pathways and proteins with related functions, including the PI3K/mTOR pathway, altered in 60% of lines; BRCA DNA repair, 44%; and SYNE1-SYNE2, 60%. Homozygous deletions of chromosome 9p21 are known to target the cell cycle regulators CDKN2A and CDKN2B. This loci was commonly lost in BCa cell lines and we show the deletions extended to the polyamine enzyme methylthioadenosine (MTA) phosphorylase (MTAP) in 36% of lines, transcription factor DMRTA1 (27%) and antiviral interferon epsilon (IFNE, 19%). Overall, the BCa cell line genomic aberrations were concordant with those found in BCa patient tumors. We used gene expression and copy number data to infer pathway activities for cell lines, then used the inferred pathway activities to build a predictive model of cisplatin response. When applied to platinum-treated patients gathered from TCGA, the model predicted treatment-specific response. Together, these data and analysis represent a valuable community resource to model basic tumor biology and to study the pharmacogenomics of BCa.
- Published
- 2016
19. A Mouse Model of Schnyder Corneal Dystrophy with the N100S Point Mutation
- Author
-
Amina S. Woods, Ludovic Muller, Michael L. Nickerson, Winston W.-Y. Kao, Jianhua Zhang, Howard S. Kruth, Michelle A. Boettler, Christian A. Combs, Xueting Jin, Harrison Sciulli, Christina Del Greco, Fei Dong, Shurong Wang, Mones Abu-Asab, Shelley N. Jackson, Jayne S. Weiss, Yueh-Chiang Hu, and Maria M Campos
- Subjects
Male ,0301 basic medicine ,Mitochondrial DNA ,lcsh:Medicine ,Biology ,Filipin ,Article ,Cornea ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Animals ,Humans ,Point Mutation ,lcsh:Science ,Corneal Dystrophies, Hereditary ,Microscopy, Confocal ,Multidisciplinary ,Point mutation ,lcsh:R ,Autosomal dominant trait ,Heterozygote advantage ,Dimethylallyltranstransferase ,Molecular biology ,Fold change ,Mitochondria ,3. Good health ,Disease Models, Animal ,Microscopy, Electron ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Glycerophosphates ,lcsh:Q ,sense organs ,CRISPR-Cas Systems ,Abnormal mitochondrial morphology - Abstract
Schnyder corneal dystrophy (SCD) is a rare autosomal dominant disease in humans, characterized by abnormal deposition of cholesterol and phospholipids in cornea caused by mutations in the UbiA prenyltransferase domain containing 1 (UBIAD1) gene. In this study, we generated a mouse line carrying Ubiad1 N100S point mutation using the CRISPR/Cas9 technique to investigate the pathogenesis of SCD. In vivo confocal microscopy revealed hyper-reflective dot-like deposits in the anterior cornea in heterozygotes and homozygotes. No significant change was found in corneal epithelial barrier function or wound healing. Electron microscopy revealed abnormal mitochondrial morphology in corneal epithelial, stromal, and endothelial cells. Mitochondrial DNA copy number assay showed 1.27 ± 0.07 fold change in homozygotes versus 0.98 ± 0.05 variation in wild type mice (P Ubiad1N100S mouse provides a promising animal model of SCD revealing that mitochondrial dysfunction is a prominent component of the disease. The different phenotype in human and mouse may due to difference in cholesterol metabolism between species.
- Published
- 2018
20. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma
- Author
-
Christopher J. Ricketts, Aguirre A. De Cubas, Huihui Fan, Christof C. Smith, Martin Lang, Ed Reznik, Reanne Bowlby, Ewan A. Gibb, Rehan Akbani, Rameen Beroukhim, Donald P. Bottaro, Toni K. Choueiri, Richard A. Gibbs, Andrew K. Godwin, Scott Haake, A. Ari Hakimi, Elizabeth P. Henske, James J. Hsieh, Thai H. Ho, Rupa S. Kanchi, Bhavani Krishnan, David J. Kwiatkowski, Wembin Lui, Maria J. Merino, Gordon B. Mills, Jerome Myers, Michael L. Nickerson, Victor E. Reuter, Laura S. Schmidt, C. Simon Shelley, Hui Shen, Brian Shuch, Sabina Signoretti, Ramaprasad Srinivasan, Pheroze Tamboli, George Thomas, Benjamin G. Vincent, Cathy D. Vocke, David A. Wheeler, Lixing Yang, William Y. Kim, A. Gordon Robertson, Paul T. Spellman, W. Kimryn Rathmell, W. Marston Linehan, Samantha J. Caesar-Johnson, John A. Demchok, Ina Felau, Melpomeni Kasapi, Martin L. Ferguson, Carolyn M. Hutter, Heidi J. Sofia, Roy Tarnuzzer, Zhining Wang, Liming Yang, Jean C. Zenklusen, Jiashan (Julia) Zhang, Sudha Chudamani, Jia Liu, Laxmi Lolla, Rashi Naresh, Todd Pihl, Qiang Sun, Yunhu Wan, Ye Wu, Juok Cho, Timothy DeFreitas, Scott Frazer, Nils Gehlenborg, Gad Getz, David I. Heiman, Jaegil Kim, Michael S. Lawrence, Pei Lin, Sam Meier, Michael S. Noble, Gordon Saksena, Doug Voet, Hailei Zhang, Brady Bernard, Nyasha Chambwe, Varsha Dhankani, Theo Knijnenburg, Roger Kramer, Kalle Leinonen, Yuexin Liu, Michael Miller, Sheila Reynolds, Ilya Shmulevich, Vesteinn Thorsson, Wei Zhang, Bradley M. Broom, Apurva M. Hegde, Zhenlin Ju, Anil Korkut, Jun Li, Han Liang, Shiyun Ling, Wenbin Liu, Yiling Lu, Kwok-Shing Ng, Arvind Rao, Michael Ryan, Jing Wang, John N. Weinstein, Jiexin Zhang, Adam Abeshouse, Joshua Armenia, Debyani Chakravarty, Walid K. Chatila, Ino de Bruijn, Jianjiong Gao, Benjamin E. Gross, Zachary J. Heins, Ritika Kundra, Konnor La, Marc Ladanyi, Augustin Luna, Moriah G. Nissan, Angelica Ochoa, Sarah M. Phillips, Francisco Sanchez-Vega, Chris Sander, Nikolaus Schultz, Robert Sheridan, S. Onur Sumer, Yichao Sun, Barry S. Taylor, Jioajiao Wang, Hongxin Zhang, Pavana Anur, Myron Peto, Paul Spellman, Christopher Benz, Joshua M. Stuart, Christopher K. Wong, Christina Yau, D. Neil Hayes, Joel S. Parker, Matthew D. Wilkerson, Adrian Ally, Miruna Balasundaram, Denise Brooks, Rebecca Carlsen, Eric Chuah, Noreen Dhalla, Robert Holt, Steven J.M. Jones, Katayoon Kasaian, Darlene Lee, Yussanne Ma, Marco A. Marra, Michael Mayo, Richard A. Moore, Andrew J. Mungall, Karen Mungall, Sara Sadeghi, Jacqueline E. Schein, Payal Sipahimalani, Angela Tam, Nina Thiessen, Kane Tse, Tina Wong, Ashton C. Berger, Andrew D. Cherniack, Carrie Cibulskis, Stacey B. Gabriel, Galen F. Gao, Gavin Ha, Matthew Meyerson, Steven E. Schumacher, Juliann Shih, Melanie H. Kucherlapati, Raju S. Kucherlapati, Stephen Baylin, Leslie Cope, Ludmila Danilova, Moiz S. Bootwalla, Phillip H. Lai, Dennis T. Maglinte, David J. Van Den Berg, Daniel J. Weisenberger, J. Todd Auman, Saianand Balu, Tom Bodenheimer, Cheng Fan, Katherine A. Hoadley, Alan P. Hoyle, Stuart R. Jefferys, Corbin D. Jones, Shaowu Meng, Piotr A. Mieczkowski, Lisle E. Mose, Amy H. Perou, Charles M. Perou, Jeffrey Roach, Yan Shi, Janae V. Simons, Tara Skelly, Matthew G. Soloway, Donghui Tan, Umadevi Veluvolu, Toshinori Hinoue, Peter W. Laird, Wanding Zhou, Michelle Bellair, Kyle Chang, Kyle Covington, Chad J. Creighton, Huyen Dinh, HarshaVardhan Doddapaneni, Lawrence A. Donehower, Jennifer Drummond, Robert Glenn, Walker Hale, Yi Han, Jianhong Hu, Viktoriya Korchina, Sandra Lee, Lora Lewis, Wei Li, Xiuping Liu, Margaret Morgan, Donna Morton, Donna Muzny, Jireh Santibanez, Margi Sheth, Eve Shinbrot, Linghua Wang, Min Wang, Liu Xi, Fengmei Zhao, Julian Hess, Elizabeth L. Appelbaum, Matthew Bailey, Matthew G. Cordes, Li Ding, Catrina C. Fronick, Lucinda A. Fulton, Robert S. Fulton, Cyriac Kandoth, Elaine R. Mardis, Michael D. McLellan, Christopher A. Miller, Heather K. Schmidt, Richard K. Wilson, Daniel Crain, Erin Curley, Johanna Gardner, Kevin Lau, David Mallery, Scott Morris, Joseph Paulauskis, Robert Penny, Candace Shelton, Troy Shelton, Mark Sherman, Eric Thompson, Peggy Yena, Jay Bowen, Julie M. Gastier-Foster, Mark Gerken, Kristen M. Leraas, Tara M. Lichtenberg, Nilsa C. Ramirez, Lisa Wise, Erik Zmuda, Niall Corcoran, Tony Costello, Christopher Hovens, Andre L. Carvalho, Ana C. de Carvalho, José H. Fregnani, Adhemar Longatto-Filho, Rui M. Reis, Cristovam Scapulatempo-Neto, Henrique C.S. Silveira, Daniel O. Vidal, Andrew Burnette, Jennifer Eschbacher, Beth Hermes, Ardene Noss, Rosy Singh, Matthew L. Anderson, Patricia D. Castro, Michael Ittmann, David Huntsman, Bernard Kohl, Xuan Le, Richard Thorp, Chris Andry, Elizabeth R. Duffy, Vladimir Lyadov, Oxana Paklina, Galiya Setdikova, Alexey Shabunin, Mikhail Tavobilov, Christopher McPherson, Ronald Warnick, Ross Berkowitz, Daniel Cramer, Colleen Feltmate, Neil Horowitz, Adam Kibel, Michael Muto, Chandrajit P. Raut, Andrei Malykh, Jill S. Barnholtz-Sloan, Wendi Barrett, Karen Devine, Jordonna Fulop, Quinn T. Ostrom, Kristen Shimmel, Yingli Wolinsky, Andrew E. Sloan, Agostino De Rose, Felice Giuliante, Marc Goodman, Beth Y. Karlan, Curt H. Hagedorn, John Eckman, Jodi Harr, Kelinda Tucker, Leigh Anne Zach, Brenda Deyarmin, Hai Hu, Leonid Kvecher, Caroline Larson, Richard J. Mural, Stella Somiari, Ales Vicha, Tomas Zelinka, Joseph Bennett, Mary Iacocca, Brenda Rabeno, Patricia Swanson, Mathieu Latour, Louis Lacombe, Bernard Têtu, Alain Bergeron, Mary McGraw, Susan M. Staugaitis, John Chabot, Hanina Hibshoosh, Antonia Sepulveda, Tao Su, Timothy Wang, Olga Potapova, Olga Voronina, Laurence Desjardins, Odette Mariani, Sergio Roman-Roman, Xavier Sastre, Marc-Henri Stern, Feixiong Cheng, Andrew Berchuck, Darell Bigner, Eric Lipp, Jeffrey Marks, Shannon McCall, Roger McLendon, Angeles Secord, Alexis Sharp, Madhusmita Behera, Daniel J. Brat, Amy Chen, Keith Delman, Seth Force, Fadlo Khuri, Kelly Magliocca, Shishir Maithel, Jeffrey J. Olson, Taofeek Owonikoko, Alan Pickens, Suresh Ramalingam, Dong M. Shin, Gabriel Sica, Erwin G. Van Meir, Hongzheng Zhang, Wil Eijckenboom, Ad Gillis, Esther Korpershoek, Leendert Looijenga, Wolter Oosterhuis, Hans Stoop, Kim E. van Kessel, Ellen C. Zwarthoff, Chiara Calatozzolo, Lucia Cuppini, Stefania Cuzzubbo, Francesco DiMeco, Gaetano Finocchiaro, Luca Mattei, Alessandro Perin, Bianca Pollo, Chu Chen, John Houck, Pawadee Lohavanichbutr, Arndt Hartmann, Christine Stoehr, Robert Stoehr, Helge Taubert, Sven Wach, Bernd Wullich, Witold Kycler, Dawid Murawa, Maciej Wiznerowicz, Ki Chung, W. Jeffrey Edenfield, Julie Martin, Eric Baudin, Glenn Bubley, Raphael Bueno, Assunta De Rienzo, William G. Richards, Steven Kalkanis, Tom Mikkelsen, Houtan Noushmehr, Lisa Scarpace, Nicolas Girard, Marta Aymerich, Elias Campo, Eva Giné, Armando López Guillermo, Nguyen Van Bang, Phan Thi Hanh, Bui Duc Phu, Yufang Tang, Howard Colman, Kimberley Evason, Peter R. Dottino, John A. Martignetti, Hani Gabra, Hartmut Juhl, Teniola Akeredolu, Serghei Stepa, Dave Hoon, Keunsoo Ahn, Koo Jeong Kang, Felix Beuschlein, Anne Breggia, Michael Birrer, Debra Bell, Mitesh Borad, Alan H. Bryce, Erik Castle, Vishal Chandan, John Cheville, John A. Copland, Michael Farnell, Thomas Flotte, Nasra Giama, Thai Ho, Michael Kendrick, Jean-Pierre Kocher, Karla Kopp, Catherine Moser, David Nagorney, Daniel O’Brien, Brian Patrick O’Neill, Tushar Patel, Gloria Petersen, Florencia Que, Michael Rivera, Lewis Roberts, Robert Smallridge, Thomas Smyrk, Melissa Stanton, R. Houston Thompson, Michael Torbenson, Ju Dong Yang, Lizhi Zhang, Fadi Brimo, Jaffer A. Ajani, Ana Maria Angulo Gonzalez, Carmen Behrens, Jolanta Bondaruk, Russell Broaddus, Bogdan Czerniak, Bita Esmaeli, Junya Fujimoto, Jeffrey Gershenwald, Charles Guo, Alexander J. Lazar, Christopher Logothetis, Funda Meric-Bernstam, Cesar Moran, Lois Ramondetta, David Rice, Anil Sood, Timothy Thompson, Patricia Troncoso, Anne Tsao, Ignacio Wistuba, Candace Carter, Lauren Haydu, Peter Hersey, Valerie Jakrot, Hojabr Kakavand, Richard Kefford, Kenneth Lee, Georgina Long, Graham Mann, Michael Quinn, Robyn Saw, Richard Scolyer, Kerwin Shannon, Andrew Spillane, onathan Stretch, Maria Synott, John Thompson, James Wilmott, Hikmat Al-Ahmadie, Timothy A. Chan, Ronald Ghossein, Anuradha Gopalan, Douglas A. Levine, Victor Reuter, Samuel Singer, Bhuvanesh Singh, Nguyen Viet Tien, Thomas Broudy, Cyrus Mirsaidi, Praveen Nair, Paul Drwiega, Judy Miller, Jennifer Smith, Howard Zaren, Joong-Won Park, Nguyen Phi Hung, Electron Kebebew, Adam R. Metwalli, Karel Pacak, Peter A. Pinto, Mark Schiffman, Nicolas Wentzensen, Robert Worrell, Hannah Yang, Marc Moncrieff, Chandra Goparaju, Jonathan Melamed, Harvey Pass, Natalia Botnariuc, Irina Caraman, Mircea Cernat, Inga Chemencedji, Adrian Clipca, Serghei Doruc, Ghenadie Gorincioi, Sergiu Mura, Maria Pirtac, Irina Stancul, Diana Tcaciuc, Monique Albert, Iakovina Alexopoulou, Angel Arnaout, John Bartlett, Jay Engel, Sebastien Gilbert, Jeremy Parfitt, Harman Sekhon, Doris M. Rassl, Robert C. Rintoul, Carlo Bifulco, Raina Tamakawa, Walter Urba, Nicholas Hayward, Henri Timmers, Anna Antenucci, Francesco Facciolo, Gianluca Grazi, Mirella Marino, Roberta Merola, Ronald de Krijger, Anne-Paule Gimenez-Roqueplo, Alain Piché, Simone Chevalier, Ginette McKercher, Kivanc Birsoy, Gene Barnett, Cathy Brewer, Carol Farver, Theresa Naska, Nathan A. Pennell, Daniel Raymond, Cathy Schilero, Kathy Smolenski, Felicia Williams, Carl Morrison, Jeffrey A. Borgia, Michael J. Liptay, Mark Pool, Christopher W. Seder, Kerstin Junker, Larsson Omberg, Mikhail Dinkin, George Manikhas, Domenico Alvaro, Maria Consiglia Bragazzi, Vincenzo Cardinale, Guido Carpino, Eugenio Gaudio, David Chesla, Sandra Cottingham, Michael Dubina, Fedor Moiseenko, Renumathy Dhanasekaran, Karl-Friedrich Becker, Klaus-Peter Janssen, Julia Slotta-Huspenina, Mohamed H. Abdel-Rahman, Dina Aziz, Sue Bell, Colleen M. Cebulla, Amy Davis, Rebecca Duell, J. Bradley Elder, Joe Hilty, Bahavna Kumar, James Lang, Norman L. Lehman, Randy Mandt, Phuong Nguyen, Robert Pilarski, Karan Rai, Lynn Schoenfield, Kelly Senecal, Paul Wakely, Paul Hansen, Ronald Lechan, James Powers, Arthur Tischler, William E. Grizzle, Katherine C. Sexton, Alison Kastl, Joel Henderson, Sima Porten, Jens Waldmann, Martin Fassnacht, Sylvia L. Asa, Dirk Schadendorf, Marta Couce, Markus Graefen, Hartwig Huland, Guido Sauter, Thorsten Schlomm, Ronald Simon, Pierre Tennstedt, Oluwole Olabode, Mark Nelson, Oliver Bathe, Peter R. Carroll, June M. Chan, Philip Disaia, Pat Glenn, Robin K. Kelley, Charles N. Landen, Joanna Phillips, Michael Prados, Jeffry Simko, Karen Smith-McCune, Scott VandenBerg, Kevin Roggin, Ashley Fehrenbach, Ady Kendler, Suzanne Sifri, Ruth Steele, Antonio Jimeno, Francis Carey, Ian Forgie, Massimo Mannelli, Michael Carney, Brenda Hernandez, Benito Campos, Christel Herold-Mende, Christin Jungk, Andreas Unterberg, Andreas von Deimling, Aaron Bossler, Joseph Galbraith, Laura Jacobus, Michael Knudson, Tina Knutson, Deqin Ma, Mohammed Milhem, Rita Sigmund, Rashna Madan, Howard G. Rosenthal, Clement Adebamowo, Sally N. Adebamowo, Alex Boussioutas, David Beer, Thomas Giordano, Anne-Marie Mes-Masson, Fred Saad, Therese Bocklage, Lisa Landrum, Robert Mannel, Kathleen Moore, Katherine Moxley, Russel Postier, Joan Walker, Rosemary Zuna, Michael Feldman, Federico Valdivieso, Rajiv Dhir, James Luketich, Edna M. Mora Pinero, Mario Quintero-Aguilo, Carlos Gilberto Carlotti, Jose Sebastião Dos Santos, Rafael Kemp, Ajith Sankarankuty, Daniela Tirapelli, James Catto, Kathy Agnew, Elizabeth Swisher, Jenette Creaney, Bruce Robinson, Carl Simon Shelley, Eryn M. Godwin, Sara Kendall, Cassaundra Shipman, Carol Bradford, Thomas Carey, Andrea Haddad, Jeffey Moyer, Lisa Peterson, Mark Prince, Laura Rozek, Gregory Wolf, Rayleen Bowman, Kwun M. Fong, Ian Yang, Robert Korst, J. Leigh Fantacone-Campbell, Jeffrey A. Hooke, Albert J. Kovatich, Craig D. Shriver, John DiPersio, Bettina Drake, Ramaswamy Govindan, Sharon Heath, Timothy Ley, Brian Van Tine, Peter Westervelt, Mark A. Rubin, Jung Il Lee, Natália D. Aredes, Armaz Mariamidze, SAIC-F-Frederick, Inc, Leidos Biomedical Research, Inc., Ricketts C.J., De Cubas A.A., Fan H., Smith C.C., Lang M., Reznik E., Bowlby R., Gibb E.A., Akbani R., Beroukhim R., Bottaro D.P., Choueiri T.K., Gibbs R.A., Godwin A.K., Haake S., Hakimi A.A., Henske E.P., Hsieh J.J., Ho T.H., Kanchi R.S., Krishnan B., Kwaitkowski D.J., Lui W., Merino M.J., Mills G.B., Myers J., Nickerson M.L., Reuter V.E., Schmidt L.S., Shelley C.S., Shen H., Shuch B., Signoretti S., Srinivasan R., Tamboli P., Thomas G., Vincent B.G., Vocke C.D., Wheeler D.A., Yang L., Kim W.T., Robertson A.G., Caesar-Johnson S.J., Demchok J.A., Felau I., Kasapi M., Ferguson M.L., Hutter C.M., Sofia H.J., Tarnuzzer R., Wang Z., Zenklusen J.C., Zhang J.J., Chudamani S., Liu J., Lolla L., Naresh R., Pihl T., Sun Q., Wan Y., Wu Y., Cho J., DeFreitas T., Frazer S., Gehlenborg N., Getz G., Heiman D.I., Kim J., Lawrence M.S., Lin P., Meier S., Noble M.S., Saksena G., Voet D., Zhang H., Bernard B., Chambwe N., Dhankani V., Knijnenburg T., Kramer R., Leinonen K., Liu Y., Miller M., Reynolds S., Shmulevich I., Thorsson V., Zhang W., Broom B.M., Hegde A.M., Ju Z., Korkut A., Li J., Liang H., Ling S., Liu W., Lu Y., Ng K.-S., Rao A., Ryan M., Wang J., Weinstein J.N., Zhang J., Abeshouse A., Armenia J., Chakravarty D., Chatila W.K., de Bruijn I., Gao J., Gross B.E., Heins Z.J., Kundra R., La K., Ladanyi M., Luna A., Nissan M.G., Ochoa A., Phillips S.M., Sanchez-Vega F., Sander C., Schultz N., Sheridan R., Sumer S.O., Sun Y., Taylor B.S., Anur P., Peto M., Spellman P.T., Benz C., Stuart J.M., Wong C.K., Yau C., Hayes D.N., Parker J.S., Wilkerson M.D., Ally A., Balasundaram M., Brooks D., Carlsen R., Chuah E., Dhalla N., Holt R., Jones S.J.M., Kasaian K., Lee D., Ma Y., Marra M.A., Mayo M., Moore R.A., Mungall A.J., Mungall K., Sadeghi S., Schein J.E., Sipahimalani P., Tam A., Thiessen N., Tse K., Wong T., Berger A.C., Cherniack A.D., Cibulskis C., Gabriel S.B., Gao G.F., Ha G., Meyerson M., Schumacher S.E., Shih J., Kucherlapati M.H., Kucherlapati R.S., Baylin S., Cope L., Danilova L., Bootwalla M.S., Lai P.H., Maglinte D.T., Van Den Berg D.J., Weisenberger D.J., Auman J.T., Balu S., Bodenheimer T., Fan C., Hoadley K.A., Hoyle A.P., Jefferys S.R., Jones C.D., Meng S., Mieczkowski P.A., Mose L.E., Perou A.H., Perou C.M., Roach J., Shi Y., Simons J.V., Skelly T., Soloway M.G., Tan D., Veluvolu U., Hinoue T., Laird P.W., Zhou W., Bellair M., Chang K., Covington K., Creighton C.J., Dinh H., Doddapaneni H., Donehower L.A., Drummond J., Glenn R., Hale W., Han Y., Hu J., Korchina V., Lee S., Lewis L., Li W., Liu X., Morgan M., Morton D., Muzny D., Santibanez J., Sheth M., Shinbrot E., Wang L., Wang M., Xi L., Zhao F., Hess J., Appelbaum E.L., Bailey M., Cordes M.G., Ding L., Fronick C.C., Fulton L.A., Fulton R.S., Kandoth C., Mardis E.R., McLellan M.D., Miller C.A., Schmidt H.K., Wilson R.K., Crain D., Curley E., Gardner J., Lau K., Mallery D., Morris S., Paulauskis J., Penny R., Shelton C., Shelton T., Sherman M., Thompson E., Yena P., Bowen J., Gastier-Foster J.M., Gerken M., Leraas K.M., Lichtenberg T.M., Ramirez N.C., Wise L., Zmuda E., Corcoran N., Costello T., Hovens C., Carvalho A.L., de Carvalho A.C., Fregnani J.H., Longatto-Filho A., Reis R.M., Scapulatempo-Neto C., Silveira H.C.S., Vidal D.O., Burnette A., Eschbacher J., Hermes B., Noss A., Singh R., Anderson M.L., Castro P.D., Ittmann M., Huntsman D., Kohl B., Le X., Thorp R., Andry C., Duffy E.R., Lyadov V., Paklina O., Setdikova G., Shabunin A., Tavobilov M., McPherson C., Warnick R., Berkowitz R., Cramer D., Feltmate C., Horowitz N., Kibel A., Muto M., Raut C.P., Malykh A., Barnholtz-Sloan J.S., Barrett W., Devine K., Fulop J., Ostrom Q.T., Shimmel K., Wolinsky Y., Sloan A.E., De Rose A., Giuliante F., Goodman M., Karlan B.Y., Hagedorn C.H., Eckman J., Harr J., Tucker K., Zach L.A., Deyarmin B., Hu H., Kvecher L., Larson C., Mural R.J., Somiari S., Vicha A., Zelinka T., Bennett J., Iacocca M., Rabeno B., Swanson P., Latour M., Lacombe L., Tetu B., Bergeron A., McGraw M., Staugaitis S.M., Chabot J., Hibshoosh H., Sepulveda A., Su T., Wang T., Potapova O., Voronina O., Desjardins L., Mariani O., Roman-Roman S., Sastre X., Stern M.-H., Cheng F., Berchuck A., Bigner D., Lipp E., Marks J., McCall S., McLendon R., Secord A., Sharp A., Behera M., Brat D.J., Chen A., Delman K., Force S., Khuri F., Magliocca K., Maithel S., Olson J.J., Owonikoko T., Pickens A., Ramalingam S., Shin D.M., Sica G., Van Meir E.G., Eijckenboom W., Gillis A., Korpershoek E., Looijenga L., Oosterhuis W., Stoop H., van Kessel K.E., Zwarthoff E.C., Calatozzolo C., Cuppini L., Cuzzubbo S., DiMeco F., Finocchiaro G., Mattei L., Perin A., Pollo B., Chen C., Houck J., Lohavanichbutr P., Hartmann A., Stoehr C., Stoehr R., Taubert H., Wach S., Wullich B., Kycler W., Murawa D., Wiznerowicz M., Chung K., Edenfield W.J., Martin J., Baudin E., Bubley G., Bueno R., De Rienzo A., Richards W.G., Kalkanis S., Mikkelsen T., Noushmehr H., Scarpace L., Girard N., Aymerich M., Campo E., Gine E., Guillermo A.L., Van Bang N., Hanh P.T., Phu B.D., Tang Y., Colman H., Evason K., Dottino P.R., Martignetti J.A., Gabra H., Juhl H., Akeredolu T., Stepa S., Hoon D., Ahn K., Kang K.J., Beuschlein F., Breggia A., Birrer M., Bell D., Borad M., Bryce A.H., Castle E., Chandan V., Cheville J., Copland J.A., Farnell M., Flotte T., Giama N., Kendrick M., Kocher J.-P., Kopp K., Moser C., Nagorney D., O'Brien D., O'Neill B.P., Patel T., Petersen G., Que F., Rivera M., Roberts L., Smallridge R., Smyrk T., Stanton M., Thompson R.H., Torbenson M., Yang J.D., Zhang L., Brimo F., Ajani J.A., Gonzalez A.M.A., Behrens C., Bondaruk J., Broaddus R., Czerniak B., Esmaeli B., Fujimoto J., Gershenwald J., Guo C., Lazar A.J., Logothetis C., Meric-Bernstam F., Moran C., Ramondetta L., Rice D., Sood A., Thompson T., Troncoso P., Tsao A., Wistuba I., Carter C., Haydu L., Hersey P., Jakrot V., Kakavand H., Kefford R., Lee K., Long G., Mann G., Quinn M., Saw R., Scolyer R., Shannon K., Spillane A., Stretch O., Synott M., Thompson J., Wilmott J., Al-Ahmadie H., Chan T.A., Ghossein R., Gopalan A., Levine D.A., Singer S., Singh B., Tien N.V., Broudy T., Mirsaidi C., Nair P., Drwiega P., Miller J., Smith J., Zaren H., Park J.-W., Hung N.P., Kebebew E., Linehan W.M., Metwalli A.R., Pacak K., Pinto P.A., Schiffman M., Wentzensen N., Worrell R., Yang H., Moncrieff M., Goparaju C., Melamed J., Pass H., Botnariuc N., Caraman I., Cernat M., Chemencedji I., Clipca A., Doruc S., Gorincioi G., Mura S., Pirtac M., Stancul I., Tcaciuc D., Albert M., Alexopoulou I., Arnaout A., Bartlett J., Engel J., Gilbert S., Parfitt J., Sekhon H., Rassl D.M., Rintoul R.C., Bifulco C., Tamakawa R., Urba W., Hayward N., Timmers H., Antenucci A., Facciolo F., Grazi G., Marino M., Merola R., de Krijger R., Gimenez-Roqueplo A.-P., Piche A., Chevalier S., McKercher G., Birsoy K., Barnett G., Brewer C., Farver C., Naska T., Pennell N.A., Raymond D., Schilero C., Smolenski K., Williams F., Morrison C., Borgia J.A., Liptay M.J., Pool M., Seder C.W., Junker K., Omberg L., Dinkin M., Manikhas G., Alvaro D., Bragazzi M.C., Cardinale V., Carpino G., Gaudio E., Chesla D., Cottingham S., Dubina M., Moiseenko F., Dhanasekaran R., Becker K.-F., Janssen K.-P., Slotta-Huspenina J., Abdel-Rahman M.H., Aziz D., Bell S., Cebulla C.M., Davis A., Duell R., Elder J.B., Hilty J., Kumar B., Lang J., Lehman N.L., Mandt R., Nguyen P., Pilarski R., Rai K., Schoenfield L., Senecal K., Wakely P., Hansen P., Lechan R., Powers J., Tischler A., Grizzle W.E., Sexton K.C., Kastl A., Henderson J., Porten S., Waldmann J., Fassnacht M., Asa S.L., Schadendorf D., Couce M., Graefen M., Huland H., Sauter G., Schlomm T., Simon R., Tennstedt P., Olabode O., Nelson M., Bathe O., Carroll P.R., Chan J.M., Disaia P., Glenn P., Kelley R.K., Landen C.N., Phillips J., Prados M., Simko J., Smith-McCune K., VandenBerg S., Roggin K., Fehrenbach A., Kendler A., Sifri S., Steele R., Jimeno A., Carey F., Forgie I., Mannelli M., Carney M., Hernandez B., Campos B., Herold-Mende C., Jungk C., Unterberg A., von Deimling A., Bossler A., Galbraith J., Jacobus L., Knudson M., Knutson T., Ma D., Milhem M., Sigmund R., Madan R., Rosenthal H.G., Adebamowo C., Adebamowo S.N., Boussioutas A., Beer D., Giordano T., Mes-Masson A.-M., Saad F., Bocklage T., Landrum L., Mannel R., Moore K., Moxley K., Postier R., Walker J., Zuna R., Feldman M., Valdivieso F., Dhir R., Luketich J., Pinero E.M.M., Quintero-Aguilo M., Carlotti C.G., Dos Santos J.S., Kemp R., Sankarankuty A., Tirapelli D., Catto J., Agnew K., Swisher E., Creaney J., Robinson B., Godwin E.M., Kendall S., Shipman C., Bradford C., Carey T., Haddad A., Moyer J., Peterson L., Prince M., Rozek L., Wolf G., Bowman R., Fong K.M., Yang I., Korst R., Rathmell W.K., Fantacone-Campbell J.L., Hooke J.A., Kovatich A.J., Shriver C.D., DiPersio J., Drake B., Govindan R., Heath S., Ley T., Van Tine B., Westervelt P., Rubin M.A., Lee J.I., Aredes N.D., and Mariamidze A.
- Subjects
Genetics and Molecular Biology (all) ,0301 basic medicine ,Transcription Factor ,Chromophobe Renal Cell Carcinoma ,papillary renal cell carcinoma ,Cell ,Cancer Genome Atlas Research Network ,Chromophobe cell ,clear cell renal cell carcinoma ,urologic and male genital diseases ,Biochemistry ,PBRM1 ,chromatin remodeling ,CDKN2A ,chromophobe renal cell carcinoma ,DNA hypermethylation ,immune signature ,PanCanAtlas ,TCGA ,0302 clinical medicine ,Renal cell carcinoma ,LS2_1 ,LS4_6 ,Biochemistry, Genetics and Molecular Biology (all) ,RNA-SEQ ,SOMATIC POINT MUTATIONS ,CLASS DISCOVERY ,lcsh:QH301-705.5 ,Nuclear Protein ,BAP1 ,Papillary renal cell carcinomas ,KIDNEY CANCER ,Kidney Neoplasm ,Nuclear Proteins ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,3. Good health ,DNA-Binding Proteins ,medicine.anatomical_structure ,Phenotype ,030220 oncology & carcinogenesis ,Survival Analysi ,MESSENGER-RNA ,Life Sciences & Biomedicine ,Ubiquitin Thiolesterase ,Metabolic Networks and Pathways ,Human ,EXPRESSION ,SEQUENCING DATA ,GENETIC-BASIS ,610 Medicine & health ,Computational biology ,Biology ,BREAST ,Article ,General Biochemistry, Genetics and Molecular Biology ,NO ,03 medical and health sciences ,Atlas (anatomy) ,Cancer genome ,medicine ,Biomarkers, Tumor ,Humans ,ESTRATÉGIAS TERAPÊUTICAS ,neoplasms ,Carcinoma, Renal Cell ,Cyclin-Dependent Kinase Inhibitor p16 ,Tumor Suppressor Protein ,Science & Technology ,Biochemistry, Genetics and Molecular Biology(all) ,Genome, Human ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Metabolic Networks and Pathway ,Cell Biology ,DNA ,medicine.disease ,Survival Analysis ,Clear cell renal cell carcinoma ,030104 developmental biology ,lcsh:Biology (General) ,Cancer research ,PanCanAtla ,Genetics and Molecular Biology(all) ,Transcription Factors - Abstract
SUMMARY Renal cell carcinoma (RCC) is not a single disease, but several histologically defined cancers with different genetic drivers, clinical courses, and therapeutic responses. The current study evaluated 843 RCC from the three major histologic subtypes, including 488 clear cell RCC, 274 papillary RCC, and 81 chromophobe RCC. Comprehensive genomic and phenotypic analysis of the RCC subtypes reveals distinctive features of each subtype that provide the foundation for the development of subtype-specific therapeutic and management strategies for patients affected with these cancers. Somatic alteration of BAP1, PBRM1, and PTEN and altered metabolic pathways correlated with subtype-specific decreased survival, while CDKN2A alteration, increased DNA hypermethylation, and increases in the immune-related Th2 gene expression signature correlated with decreased survival within all major histologic subtypes. CIMP-RCC demonstrated an increased immune signature, and a uniform and distinct metabolic expression pattern identified a subset of metabolically divergent (MD) ChRCC that associated with extremely poor survival., Graphical abstract In Brief Ricketts et al. find distinctive features of each RCC subtype, providing the foundation for development of subtypespecific therapeutic and management strategies. Somatic alteration of BAP1, PBRM1, and metabolic pathways correlates with subtype-specific decreased survival, while CDKN2A alteration, DNA hypermethylation, and Th2 immune signature correlate with decreased survival within all subtypes.
- Published
- 2017
21. The centers of premelton signals, the beginning and ends of genes
- Author
-
Michael L Nickerson
- Subjects
Schnyder corneal dystrophy ,chemistry.chemical_compound ,Biochemistry ,chemistry ,Cholesterol ,Prenyltransferase ,Dystrophy ,Cholesterol metabolism ,Metabolism ,Biology ,Function (biology) - Published
- 2017
22. Identification of novel mutations by exome sequencing in African American colorectal cancer patients
- Author
-
Hassan Ashktorab, Mohammad Daremipouran, Hassan Brim, Hamed Rahi, Edward Lee, Russell Schwartz, Joe Devaney, Michael L. Nickerson, Sudhir Varma, and Babak Shokrani
- Subjects
Genetics ,Sanger sequencing ,Cancer Research ,education.field_of_study ,dbSNP ,Population ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,symbols.namesake ,Oncology ,medicine ,symbols ,KRAS ,1000 Genomes Project ,International HapMap Project ,education ,neoplasms ,Exome sequencing - Abstract
BACKGROUND The purpose of this study was to identify genome-wide single nucleotide variants and mutations in African American patients with colorectal cancer (CRC). There is a need of such studies in African Americans, because they display a higher incidence of aggressive CRC tumors. METHODS We performed whole exome sequencing (WES) on DNA from 12 normal/tumor pairs of African American CRC patient tissues. Data analysis was performed using the software package GATK (Genome Analysis Tool Kit). Normative population databases (eg, 1000 Genomes SNP database, dbSNP, and HapMap) were used for comparison. Variants were annotated using analysis of variance and were validated via Sanger sequencing. RESULTS We identified somatic mutations in genes that are known targets in CRC such as APC, BRAF, KRAS, and PIK3CA. We detected novel alterations in the Wnt pathway gene, APC, within its exon 15, of which mutations are highly associated with CRC. CONCLUSIONS This WES study in African American patients with CRC provides insight into the identification of novel somatic mutations in APC. Our data suggest an association between specific mutations in the Wnt signaling pathway and an increased risk of CRC. The analysis of the pathogenicity of these novel variants may shed light on the aggressive nature of CRC in African Americans. Cancer 2015;121:34–42. © 2014 American Cancer Society.
- Published
- 2014
23. Concurrent Alterations in TERT, KDM6A, and the BRCA Pathway in Bladder Cancer
- Author
-
Dan Theodorescu, M. Scott Lucia, Shirley Tsang, Kate M. Im, Christina T. Ruiz-Rodriguez, Garrett M. Dancik, Lee E. Moore, Sevilay Turan, Yingrui Li, Joseph Brown, Quan Zhou, Michael G. Edwards, Michael Dean, Charles Owens, Guangwu Guo, Michael L. Nickerson, James C. Costello, and Zhiming Cai
- Subjects
Cancer Research ,Telomerase ,Biology ,Article ,Transcriptome ,medicine ,Humans ,Exome ,Exome sequencing ,Histone Demethylases ,Carcinoma, Transitional Cell ,BAP1 ,Bladder cancer ,BRCA1 Protein ,Tumor Suppressor Proteins ,Nuclear Proteins ,Cancer ,medicine.disease ,Molecular biology ,Urinary Bladder Neoplasms ,Oncology ,Mutation ,Cancer cell ,Cancer research ,Ubiquitin Thiolesterase - Abstract
Purpose: Genetic analysis of bladder cancer has revealed a number of frequently altered genes, including frequent alterations of the telomerase (TERT) gene promoter, although few altered genes have been functionally evaluated. Our objective is to characterize alterations observed by exome sequencing and sequencing of the TERT promoter, and to examine the functional relevance of histone lysine (K)–specific demethylase 6A (KDM6A/UTX), a frequently mutated histone demethylase, in bladder cancer. Experimental Design: We analyzed bladder cancer samples from 54 U.S. patients by exome and targeted sequencing and confirmed somatic variants using normal tissue from the same patient. We examined the biologic function of KDM6A using in vivo and in vitro assays. Results: We observed frequent somatic alterations in BRCA1 associated protein-1 (BAP1) in 15% of tumors, including deleterious alterations to the deubiquitinase active site and the nuclear localization signal. BAP1 mutations contribute to a high frequency of tumors with breast cancer (BRCA) DNA repair pathway alterations and were significantly associated with papillary histologic features in tumors. BAP1 and KDM6A mutations significantly co-occurred in tumors. Somatic variants altering the TERT promoter were found in 69% of tumors but were not correlated with alterations in other bladder cancer genes. We examined the function of KDM6A, altered in 24% of tumors, and show depletion in human bladder cancer cells, enhanced in vitro proliferation, in vivo tumor growth, and cell migration. Conclusions: This study is the first to identify frequent BAP1 and BRCA pathway alterations in bladder cancer, show TERT promoter alterations are independent of other bladder cancer gene alterations, and show KDM6A loss is a driver of the bladder cancer phenotype. Clin Cancer Res; 20(18); 4935–48. ©2014 AACR.
- Published
- 2014
24. Abstract LB-051: Integrative, targeted deep sequencing of bladder tumors reveals novel associations between cancer gene mutations and mutational signatures with major risk factors
- Author
-
Stella Koutros, Nina Rao, Lee E. Moore, Michael L. Nickerson, Donghyuk Lee, Bin Zhu, Larissa Pardo, Dalsu Baris, Molly Schwenn, Alison Johnson, Kristine Jones, Montserrat Garcia-Closas, Ludmila Prokunina-Olsson, Debra T. Silverman, Nathaniel Rothman, and Michael Dean
- Subjects
Cancer Research ,Oncology - Abstract
Exome and whole-genome sequencing of muscle-invasive (MI) bladder cancer has revealed important insights into the molecular landscape; however, there are few studies of non-muscle invasive (NMI) bladder cancer with detailed information on bladder cancer risk factors. We used deep, targeted amplicon sequencing of the coding regions of 44 genes frequently mutated in bladder cancer (average 630x coverage for tumor and 350x for germline DNA) to characterize somatic mutations and the mutational spectrum in 307 formalin fixed paraffin embedded bladder tumors (n=260 NMI and n=47 MI) from a population-based case-control study with detailed information on smoking and other risk factors. Variants identified by mutation calling pipelines were validated by manual review of aligned sequences. Logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) to evaluate somatic mutations and risk factors. We used SignatureEstimation to extract the four known single base substitution mutational signatures (COSMIC: Sig1, Sig2, Sig13, ERCC2-Signature) and Poisson regression to calculate risk ratios (RRs) and 95%CIs to evaluate signatures and risk factors. The most frequently mutated gene in NMI tumors was FGFR3 (52% mutated) and in MI tumors was TP53 (51% mutated). FGFR3 (89%), STAG2 (81%), and PIK3CA (78%) mutations were significantly higher in Ta tumors compared to T1 or MI tumors. Mutations in histone/chromatin regulating genes (KDM6A, KMT2D, KMT2C, CREBBP, ARID1A, and EP300) were present at a high frequency in low grade Ta tumors. Non-silent KDM6Amutations were significantly more common in females compared to males (OR=1.79,95%CI:1.03-3.11); in females these mutations largely occurred in Ta tumors (86%). There was striking heterogeneity in the relationship between smoking status (current, former, never) and the established signatures: only current smoking was associated with greater ERCC2-Signature variants compared to both former smoking (p-value=0.015) and never smoking (p-value=0.023); only former smoking was associated with APOBEC-Sig13 variants (RR=1.83,95%CI: 1.10-3.04,p-value=0.019); and only never smoking was associated with the APOBEC-Sig2 variants (RR=1.52,95%CI: 1.15-2.01,p-value=0.003). Further, there was significant evidence that smoking duration, the component of smoking most strongly associated with bladder cancer risk, was associated with ERCC2-Signature variants and APOBEC-Sig13 variants among current (p-trend=0.013) and former smokers (p=0.002), respectively. Exploration of insertion/deletions revealed that both status and duration of smoking was significantly associated with presence of any 1bp deletions (p-trend=0.007); these were enriched for deletions in KDM6A (p=0.0003). In conclusion, these data quantify the contribution of bladder cancer risk factors to mutational burden in a population-based series of tumors and reveal distinct signature contributions among never, former and current smokers. Citation Format: Stella Koutros, Nina Rao, Lee E. Moore, Michael L. Nickerson, Donghyuk Lee, Bin Zhu, Larissa Pardo, Dalsu Baris, Molly Schwenn, Alison Johnson, Kristine Jones, Montserrat Garcia-Closas, Ludmila Prokunina-Olsson, Debra T. Silverman, Nathaniel Rothman, Michael Dean. Integrative, targeted deep sequencing of bladder tumors reveals novel associations between cancer gene mutations and mutational signatures with major risk factors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-051.
- Published
- 2019
25. The TERE1 protein interacts with mitochondrial TBL2: Regulation of trans-membrane potential, ROS/RNS and SXR target genes
- Author
-
S. Bruce Malkowicz, Li-Ping Wang, Yongmu Zheng, Frank J. RauscherIII, Michael L. Nickerson, Howard S. Kruth, William J. Fredericks, Robert J. Lee, Nathaniel J. Fredericks, Huiyi Wang, Hankun Yin, Terry McGarvey, Wayland Hsiao, and Jayne S. Weiss
- Subjects
Endoplasmic reticulum ,Cell Biology ,Oxidative phosphorylation ,Golgi apparatus ,Mitochondrion ,Biology ,medicine.disease_cause ,Biochemistry ,Cell biology ,symbols.namesake ,Nuclear receptor ,symbols ,medicine ,Ectopic expression ,Signal transduction ,Molecular Biology ,Oxidative stress - Abstract
We originally discovered TERE1 as a potential tumor suppressor protein based upon reduced expression in bladder and prostate cancer specimens and growth inhibition of tumor cell lines/xenografts upon ectopic expression. Analysis of TERE1 (aka UBIAD1) has shown it is a prenyltransferase enzyme in the natural bio-synthetic pathways for both vitamin K-2 and COQ10 production and exhibits multiple subcellular localizations including mitochondria, endoplasmic reticulum, and golgi. Vitamin K-2 is involved in mitochondrial electron transport, SXR nuclear hormone receptor signaling and redox cycling: together these functions may form the basis for tumor suppressor function. To gain further insight into mechanisms of growth suppression and enzymatic regulation of TERE1 we isolated TERE1 associated proteins and identified the WD40 repeat, mitochondrial protein TBL2. We examined whether disease specific mutations in TERE1 affected interactions with TBL2 and the role of each protein in altering mitochondrial function, ROS/RNS production and SXR target gene regulation. Biochemical binding assays demonstrated a direct, high affinity interaction between TERE1 and TBL2 proteins; TERE1 was localized to both mitochondrial and non-mitochondrial membranes whereas TBL2 was predominantly mitochondrial; multiple independent single amino acid substitutions in TERE1 which cause a human hereditary corneal disease reduced binding to TBL2 strongly suggesting the relevance of this interaction. Ectopic TERE1 expression elevated mitochondrial trans-membrane potential, oxidative stress, NO production, and activated SXR targets. A TERE1-TBL2 complex likely functions in oxidative/nitrosative stress, lipid metabolism, and SXR signaling pathways in its role as a tumor suppressor.
- Published
- 2013
26. Targeted exome sequencing reveals distinct pathogenic variants in Iranians with colorectal cancer
- Author
-
Michael L. Nickerson, Hasti Olumi, Hamed Azimi, Hassan Ashktorab, Pooneh Mokarram, Hassan Brim, and Sudhir Varma
- Subjects
0301 basic medicine ,Colorectal cancer ,Population ,targeted exome sequencing ,Iran ,Caucasian ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Predictive Value of Tests ,Databases, Genetic ,Exome Sequencing ,medicine ,Biomarkers, Tumor ,Exome ,Genetic Predisposition to Disease ,education ,neoplasms ,Exome sequencing ,Genetics ,education.field_of_study ,colon ,business.industry ,Gene Expression Profiling ,Shirazi ,Cancer ,Computational Biology ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Ion semiconductor sequencing ,medicine.disease ,Prognosis ,digestive system diseases ,3. Good health ,MSH6 ,030104 developmental biology ,Phenotype ,Oncology ,MSH3 ,030220 oncology & carcinogenesis ,KRAS ,business ,Colorectal Neoplasms ,Transcriptome ,Iranian ,Research Paper - Abstract
// Hassan Ashktorab 1 , Pooneh Mokarram 5 , Hamed Azimi 1 , Hasti Olumi 1 , Sudhir Varma 3 , Michael L. Nickerson 4 , Hassan Brim 2 1 Department of Medicine and Cancer Center, Howard University College of Medicine, Washington, DC, USA 2 Department of Pathology, Howard University College of Medicine, Washington, DC, USA 3 Hithru LLC, Silver Spring, MD, USA 4 Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD, USA 5 Current address: Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran Correspondence to: Hassan Ashktorab, email: hashktorab@howard.edu Michael L. Nickerson, email: nickersonml@od.nih.gov Keywords: targeted exome sequencing, colon, Iranian, Shirazi, Caucasian Received: July 20, 2016 Accepted: December 01, 2016 Published: December 16, 2016 ABSTRACT PURPOSE: Next Generation Sequencing (NGS) is currently used to establish mutational profiles in many multigene diseases such as colorectal cancer (CRC), which is on the rise in many parts of the developing World including, Iran. Little is known about its genetic hallmarks in these populations. AIM: To identify variants in 15 CRC-associated genes in patients of Iranian descent. RESULTS: There were 51 validated variants distributed on 12 genes: 22% MSH3 ( n = 11/51), 10% MSH6 ( n = 5/51), 8% AMER1 ( n = 4/51), 20% APC ( n = 10/51), 2% BRAF ( n = 1/51), 2% KRAS ( n = 1/51), 12% PIK3CA ( n = 6/51), 8% TGFβR2A ( n = 4/51), 2% SMAD4 ( n = 1/51), 4% SOX9 ( n = 2/51), 6% TCF7L2 ( n = 3/51), and 6% TP53 ( n = 3/51). Most known and distinct variants were in mismatch repair genes ( MMR , 32%) and APC (20%). Among oncogenes, PIK3CA was the top target (12%). MATERIALS AND METHODS: CRC specimens from 63 Shirazi patients were used to establish the variant’ profile on an Ion Torrent platform by targeted exome sequencing. To rule-out technical artifacts, the variants were validated in 13 of these samples using an Illumina NGS platform. Validated variants were annotated and compared to variants from publically available databases. An in-silico functional analysis was performed. MSI status of the analyzed samples was established. CONCLUSION: These results illustrate for the first time CRC mutational profile in Iranian patients. MSH3 , MSH6 , APC and PIK3CA genes seem to play a bigger role in the path to cancer in this population. These findings will potentially lead to informed genetic diagnosis protocol and targeted therapeutic strategies.
- Published
- 2016
27. Targeted Exome Sequencing Outcome Variations of Colorectal Tumors within and across Two Sequencing Platforms
- Author
-
Hassan Ashktorab, Sara Bass, Hassan Brim, Sudhir Varma, Michael L. Nickerson, and Hamed Azimi
- Subjects
Sanger sequencing ,symbols.namesake ,Concordance ,symbols ,Ion semiconductor sequencing ,Biology ,Exome ,Molecular biology ,DNA sequencing ,Exome sequencing ,Illumina dye sequencing ,Article ,Colorectal Tumors - Abstract
Background and Aim: Next generation sequencing (NGS) has quickly the tool of choice for genome and exome data generation. The multitude of sequencing platforms as well as the variabilities within each platform need to be assessed. In this paper we used two platforms (Ion Torrent and Illumina) to assess single nucleotides variants in colorectal cancer (CRC) specimens. Methods: CRC specimens (n = 13) collected from 6 CRC (cancer and matched normal) patients were used to establish the mutational profile using Ion Torrent and Illumina sequencing platforms. We analyzed a set of samples from Formalin Fixed Paraffin Embedded and Fresh Frozen (FF) samples on both platforms to assess the effect of sample nature (FFPE vs. FF) on sequencing outcome and to evaluate the similarity/differences of SNVs across the two platforms. In addition, duplicates of fresh frozen samples were sequenced on each platform to assess variability within platform. Results: The comparison of fresh frozen replicates to each other gave a concordance of 77% (± 15.3%) in Ion Torrent and 70% (± 3.7%) in Illumina. FFPE vs. Fresh Frozen replicates gave a concordance of 40% (± 32%) in Ion Torrent and 49% (± 19%) in Illumina. For the cross platform concordance were FFPE compared to fresh frozen (Average of 75% (± 9.8%) for FFPE samples and 67% (± 32%) for fresh frozen and 70% (± 26.8%) overall average). Conclusion: Our data show a significant variability within and across platforms. Also the number of detected variants depend on the nature of the specimen; fresh frozen vs. FFPE. Validation of NGS discovered mutations is a must to rule-out false positive mutants. This validation might either be performed through a second NGS platform or through Sanger sequencing.
- Published
- 2016
28. The UBIAD1 Prenyltransferase Links Menaquione-4 Synthesis to Cholesterol Metabolic Enzymes
- Author
-
Allen D. Bosley, Thorkell Andresson, Michael L. Nickerson, Yoshihisa Hirota, Howard S. Kruth, Shigeru Kinoshita, Dominic Esposito, Jayne S. Weiss, Scott G. Morham, Wolfgang Brandt, Ludger A. Wessjohann, Brittany N. Kostiha, Michael Dean, and Toshio Okano
- Subjects
chemistry.chemical_classification ,SOAT1 ,Immunoprecipitation ,Cholesterol ,Autosomal dominant trait ,Biology ,medicine.disease ,Transmembrane domain ,chemistry.chemical_compound ,Enzyme ,Biochemistry ,chemistry ,Genetics ,Schnyder crystalline corneal dystrophy ,medicine ,Missense mutation ,Genetics (clinical) - Abstract
Schnyder corneal dystrophy (SCD) is an autosomal dominant disease characterized by germline variants in UBIAD1 introducing missense alterations leading to deposition of cholesterol in the cornea, progressive opacification, and loss of visual acuity. UBIAD1 was recently shown to synthesize menaquinone-4 (MK-4, vitamin K(2) ), but causal mechanisms of SCD are unknown. We report a novel c.864G>A UBIAD1 mutation altering glycine 177 to glutamic acid (p.G177E) in six SCD families, including four families from Finland who share a likely founder mutation. We observed reduced MK-4 synthesis by UBIAD1 altered by SCD mutations p.N102S, p.G177R/E, and p.D112N, and molecular models showed p.G177-mutant UBIAD1 disrupted transmembrane helices and active site residues. We show UBIAD1 interacts with HMGCR and SOAT1, enzymes catalyzing cholesterol synthesis and storage, respectively, using yeast two-hybrid screening and immunoprecipitation. Docking simulations indicate cholesterol binds to UBIAD1 in the substrate-binding cleft and substrate-binding overlaps with GGPP binding, an MK-4 substrate, suggesting potential competition between these metabolites. Impaired MK-4 synthesis is a biochemical defect identified in SCD suggesting UBIAD1 links vitamin K and cholesterol metabolism through physical contact between enzymes and metabolites. Our data suggest a role for endogenous MK-4 in maintaining cornea health and visual acuity.
- Published
- 2012
29. Inactivation of the von Hippel–Lindau tumor suppressor leads to selective expression of a human endogenous retrovirus in kidney cancer
- Author
-
Michael I. Lerman, Elena Cherkasova, Xin Tian, Ramaprasad Srinivasan, Julie Hong, David S. Schrump, W. M. Linehan, V. N. Senchenko, Elizabeth B Malinzak, Sheila Rao, Michael L. Nickerson, Maria J. Merino, Richard W. Childs, Anna V. Kudryavtseva, and Yoshiyuki Takahashi
- Subjects
renal cell carcinoma ,Cancer Research ,Small interfering RNA ,Tumor suppressor gene ,viruses ,Endogenous retrovirus ,urologic and male genital diseases ,Article ,Proviruses ,endogenous retrovirus ,Cell Line, Tumor ,VHL ,Von Hippel–Lindau tumor suppressor ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,Humans ,HIF ,Promoter Regions, Genetic ,Carcinoma, Renal Cell ,Molecular Biology ,DNA methylation ,biology ,Endogenous Retroviruses ,Terminal Repeat Sequences ,Transfection ,Molecular biology ,Kidney Neoplasms ,Long terminal repeat ,Von Hippel-Lindau Tumor Suppressor Protein ,biology.protein ,5' Untranslated Regions ,Chromatin immunoprecipitation - Abstract
A human endogenous retrovirus type E (HERV-E) was recently found to be selectively expressed in most renal cell carcinomas (RCCs). Importantly, antigens derived from this provirus are immunogenic, stimulating cytotoxic T cells that kill RCC cells in vitro and in vivo. Here, we show HERV-E expression is restricted to the clear cell subtype of RCC (ccRCC) characterized by an inactivation of the von Hippel-Lindau (VHL) tumor-suppressor gene with subsequent stabilization of hypoxia-inducible transcription factors (HIFs)-1α and -2α. HERV-E expression in ccRCC linearly correlated with HIF-2α levels and could be silenced in tumor cells by either transfection of normal VHL or small interfering RNA inhibition of HIF-2α. Using chromatin immunoprecipitation, we demonstrated that HIF-2α can serve as transcriptional factor for HERV-E by binding with HIF response element (HRE) localized in the proviral 5' long terminal repeat (LTR). Remarkably, the LTR was found to be hypomethylated only in HERV-E-expressing ccRCC while other tumors and normal tissues possessed a hypermethylated LTR preventing proviral expression. Taken altogether, these findings provide the first evidence that inactivation of a tumor suppressor gene can result in aberrant proviral expression in a human tumor and give insights needed for translational research aimed at boosting human immunity against antigenic components of this HERV-E.
- Published
- 2011
30. Abstract 4237: Differences in the frequencies of tumor VHL mutation and HIF-2α expression between black and white patients with clear cell renal carcinoma
- Author
-
Lee E. Moore, Maria Merino, Julie J. Ruterbusch, Faith G. Davis, Jonathan N. Hofmann, Catherine L. Callahan, Michael L. Nickerson, Stephen M. Hewitt, Kendra Schwartz, Mark P. Purdue, W. Marston Linehan, Petra Lenz, Nathaniel Rothman, and Wong-Ho Chow
- Subjects
White (mutation) ,Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,Expression (architecture) ,Clear Cell Renal Carcinoma ,medicine ,Biology - Abstract
Background: Black Americans have a poorer prognosis for clear cell renal cell carcinoma (ccRCC) than white Americans, potentially due in part to differences in tumor biology. In a recent analysis of The Cancer Genome Atlas (TCGA), tumors from black ccRCC patients had a lower rate of mutation in the VHL tumor suppressor gene and lower expression of hypoxia inducible factors (HIF) than tumors from white ccRCC patients. However, as this study was based on a small number of black patients (N=19) and had limited information on patients' medical histories and risk factors profiles, further investigation is needed. Objective: We evaluated differences in the frequencies of somatic VHL gene mutations and HIF-1α and -2α protein expression between tumors from black and white ccRCC patients. Methods: The investigation utilized formalin-fixed tissue and data collected from patients participating in a case-control study conducted in Chicago and Detroit. We sequenced VHL using the Ion Torrent platform for tumors from 69 black and 98 white patients, and measured tumor HIF-1α and -2α protein expression for 88 black and 240 white patients using immunohistochemistry. Results: Black patients' tumors had a lower frequency of VHL mutation than those of white patients (32% vs. 49%; P = 0.03) as well as a lower frequency of above-median HIF-2α expression (33% vs. 56%; P=0.002). HIF-1α expression did not differ by race (P=0.14). These racial differences persisted after multivariable model adjustment for age, sex, hypertension, chronic kidney disease, body mass index, smoking status, stage, grade, and tumor size [VHL mutation: odds ratio (OR) = 0.44, 95% confidence interval (CI) = 0.19, 0.98; HIF-2α expression: OR = 0.33, 95% CI = 0.18, 0.61]. Conclusions: Our observation that VHL mutation and high HIF-2α expression are less frequent in ccRCC tumors of black vs. white patients confirms the earlier TCGA finding. These findings suggest that ccRCC in black patients is a fundamentally different disease than ccRCC in white patients. Citation Format: Catherine L. Callahan, Lee E. Moore, Petra Lenz, Kendra Schwartz, Julie Ruterbusch, Faith Davis, Wong-Ho Chow, W. Marston Linehan, Maria J. Merino, Stephen M. Hewitt, Nathaniel Rothman, Jonathan N. Hofmann, Michael L. Nickerson, Mark P. Purdue. Differences in the frequencies of tumor VHL mutation and HIF-2α expression between black and white patients with clear cell renal carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4237.
- Published
- 2018
31. miR-23b* targets proline oxidase, a novel tumor suppressor protein in renal cancer
- Author
-
Lucy M. Anderson, Hongshan Wang, Yih-Horng Shiao, Alan O. Perantoni, Michael L. Nickerson, James M. Phang, Zabirnyk O, Khalil S, and Wen-Bin Liu
- Subjects
Regulation of gene expression ,Cancer Research ,Tumor suppressor gene ,Oncogene ,Proline oxidase ,viruses ,virus diseases ,Biology ,medicine.disease_cause ,Cancer cell ,Immunology ,microRNA ,Genetics ,Cancer research ,medicine ,Gene silencing ,Carcinogenesis ,Molecular Biology - Abstract
Proline oxidase (POX) is a novel mitochondrial tumor suppressor that can suppress proliferation and induce apoptosis through the generation of reactive oxygen species (ROS) and decreasing hypoxia-inducible factor (HIF) signaling. Recent studies have shown the absence of expression of POX in human cancer tissues, including renal cancer. However, the mechanism for the loss of POX remains obscure. No genetic or epigenetic variation of POX gene was found. In this study, we identified the upregulated miR-23b in renal cancer as an important regulator of POX. Ectopic overexpression of miR-23b in normal renal cells resulted in striking downregulation of POX, whereas POX expression increased markedly when endogenous miR-23b was knocked down by its antagomirs in renal cancer cells. Consistent with the POX-mediated tumor suppression pathway, these antagomirs induced ROS, inhibited HIF signaling and increased apoptosis. Furthermore, we confirmed the regulation of miR-23b on POX and its function in the DLD1 Tet-off POX cell system. Using a luciferase reporter system, we verified the direct binding of miR-23b to the POX mRNA 3'-untranslated region. In addition, pairs of human renal carcinoma and normal tissues showed a negative correlation between miR-23b and POX protein expression, providing its clinical corroboration. Taken together, our results suggested that miR-23b, by targeting POX, could function as an oncogene; decreasing miR-23b expression may prove to be an effective way of inhibiting kidney tumor growth.
- Published
- 2010
32. Genetic analysis of 14 families with Schnyder crystalline corneal dystrophy reveals clues to UBIAD1 protein function
- Author
-
William J. Dupps, Walter Lisch, Gerard Tromp, Peter White, Chaesik Kim, Neil D. Ebenezer, Christopher J. Rapuano, Helena Kuivaniemi, Jayaprakash Karkera, Fung-Rong Hu, Jayne S. Weiss, James J. Reidy, Da Wen Lu, Michael L. Nickerson, R. Scott Winters, Howard S. Kruth, Sunil Mahurkar, and John E. Sutphin
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,DNA Mutational Analysis ,Molecular Sequence Data ,Corneal dystrophy ,Biology ,medicine.disease_cause ,Genetic analysis ,DNA sequencing ,chemistry.chemical_compound ,Exon ,Apolipoproteins E ,Genetics ,medicine ,Humans ,Point Mutation ,Family ,Amino Acid Sequence ,Child ,Genetics (clinical) ,Aged ,Aged, 80 and over ,Corneal Dystrophies, Hereditary ,Mutation ,Protein function ,Autosomal dominant trait ,Proteins ,Middle Aged ,Dimethylallyltranstransferase ,medicine.disease ,Pedigree ,chemistry ,Amino Acid Substitution ,Schnyder crystalline corneal dystrophy ,Medical genetics ,Female ,DNA ,Protein Binding - Abstract
Schnyder crystalline corneal dystrophy (SCCD) is a rare autosomal dominant disease characterized by progressive corneal opacification resulting from abnormal deposition of cholesterol and phospholipids. Recently, six different mutations on the UBIAD1 gene on chromosome 1p36 were found to result in SCCD. The purpose of this article is to further characterize the mutation spectrum of SCCD and identify structural and functional consequences for UBIAD1 protein activity. DNA sequencing was performed on samples from 36 individuals from 14 SCCD families. One affected individual was African American and SCCD has not been previously reported in this ethnic group. We identified UBIAD1 mutations in all 14 families which had 30 affected and 6 unaffected individuals. Eight different UBIAD1 mutations, 5 novel (L121F, D118G, and S171P in exon 1, G186R and D236E in exon 2) were identified. In four families with DNA samples from both affected and unaffected individuals, the D118G, G186R, T175I, and G177R mutations cosegregated with SCCD. In combination with our previous report, we have identified the genetic mutation in UBIAD1 in 20 unrelated families with 10 (including 5 reported here), having the N102S mutation. The results suggest that N102S may be a mutation hot spot because the affected families were unrelated including Caucasian and Asian individuals. There was no genotype phenotype correlation except for the T175I mutation which demonstrated prominent diffuse corneal haze, typically without corneal crystals. Protein analysis revealed structural and functional implications of SCCD mutations which may affect UBIAD1 function, ligand binding and interaction with binding partners, like apo E.
- Published
- 2008
33. TET2 binds the androgen receptor and loss is associated with prostate cancer
- Author
-
K Misner, Zhao-Qi Wang, Lee E. Moore, Thorkell Andresson, Sonja I. Berndt, Stephen K. Anderson, A Esnakula, Jonathan R. Keller, Wen Yi Huang, Hong Lou, Joseph Boland, D Butcher, Sevilay Turan, Tongwu Zhang, Margaret A. Tucker, Jean-Noel Billaud, Seth A. Brodie, Meredith Yeager, S. J. Chanock, Kevin M. Brown, K M Im, W Tan, W. Isaacs, Dominic Esposito, Aaron J. Bouk, Hongchuan Li, Michael L. Nickerson, Shiva K. Das, Robert N. Hoover, Lei Sun, and Michael Dean
- Subjects
0301 basic medicine ,Male ,Cancer Research ,cancer risk variant ,oxoglutarate ,SDHA ,Biology ,androgen signaling ,Polymorphism, Single Nucleotide ,Article ,Dioxygenases ,03 medical and health sciences ,Prostate cancer ,Prostate ,metabolic sensor ,Proto-Oncogene Proteins ,LNCaP ,Genetics ,medicine ,Humans ,Molecular Biology ,Cell Proliferation ,BAP1 ,Gene knockdown ,epigenetics ,Prostatic Neoplasms ,Succinates ,Cell cycle ,Prostate-Specific Antigen ,medicine.disease ,Introns ,Androgen receptor ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Receptors, Androgen ,Cancer research ,Ketoglutaric Acids ,Kallikreins - Abstract
Genetic alterations associated with prostate cancer (PCa) may be identified by sequencing metastatic tumor genomes to identify molecular markers at this lethal stage of disease. Previously, we characterized somatic alterations in metastatic tumors in the methylcytosine dioxygenase ten-eleven translocation 2 (TET2), which is altered in 5–15% of myeloid, kidney, colon and prostate cancers. Genome-wide association studies previously identified non-coding risk variants associated with PCa and melanoma. We performed fine-mapping of PCa risk across TET2 using genotypes from the PEGASUS case-control cohort and identified six new risk variants in introns 1 and 2. Oligonucleotides containing two risk variants were bound by the transcription factor octamer-binding protein 1 (Oct1/POU2F1) and TET2 and Oct1 expression were positively correlated in prostate tumors. TET2 is expressed in normal prostate tissue and reduced in a subset of tumors from the Cancer Genome Atlas (TCGA). Small interfering RNA (siRNA)-mediated TET2 knockdown (KD) increases LNCaP cell proliferation, migration, and wound healing, verifying loss drives a cancer phenotype. Endogenous TET2 bound the androgen receptor (AR) and AR-coactivator proteins in LNCaP cell extracts, and TET2 KD increases prostate-specific antigen (KLK3/PSA) expression. Published data reveal TET2 binding sites and hydroxymethylcytosine (hmC) proximal to KLK3. A gene co-expression network identified using TCGA prostate tumor RNA-sequencing identifies co-regulated cancer genes associated with 2-oxoglutarate (2-OG) and succinate metabolism, including TET2, lysine demethylase (KDM) KDM6A, BRCA1-associated BAP1, and citric acid cycle enzymes IDH1/2, SDHA/B, and FH. The co-expression signature is conserved across 31 TCGA cancers suggesting a putative role for TET2 as an energy sensor (of 2-OG) that modifies aspects of androgen-AR signaling. Decreased TET2 mRNA expression in TCGA PCa tumors is strongly associated with reduced patient survival indicating reduced expression in tumors maybe an informative biomarker of disease progression and perhaps metastatic disease.
- Published
- 2015
34. The genome of Diuraphis noxia, a global aphid pest of small grains
- Author
-
Scott J. Nicholson, Yan Song, Changhoon Kim, Michael Dean, Peter R. Hoyt, Hwanseok Rhee, Michael L. Nickerson, and Gary J. Puterka
- Subjects
Insecticides ,Genotype ,Genetic Linkage ,Genome, Insect ,Drug Resistance ,Genomics ,Russian wheat aphid ,Genome ,Diuraphis noxia ,Epigenesis, Genetic ,Cytosine ,Glucose dehydrogenase ,Genetics ,Animals ,Phytotoxic ,Sugar transporter ,Gene ,Aphid ,Phylogeny ,Repetitive Sequences, Nucleic Acid ,Comparative genomics ,Base Composition ,biology ,food and beverages ,Computational Biology ,DNA Methylation ,biology.organism_classification ,Acyrthosiphon pisum ,Insect Vectors ,Aphids ,Plant-insect interactions ,DNA Transposable Elements ,Insect Proteins ,RNA Interference ,Biotechnology ,Signal Transduction ,Research Article - Abstract
Background The Russian wheat aphid, Diuraphis noxia Kurdjumov, is one of the most important pests of small grains throughout the temperate regions of the world. This phytotoxic aphid causes severe systemic damage symptoms in wheat, barley, and other small grains as a direct result of the salivary proteins it injects into the plant while feeding. Results We sequenced and de novo assembled the genome of D. noxia Biotype 2, the strain most virulent to resistance genes in wheat. The assembled genomic scaffolds span 393 MB, equivalent to 93% of its 421 MB genome, and contains 19,097 genes. D. noxia has the most AT-rich insect genome sequenced to date (70.9%), with a bimodal CpG(O/E) distribution and a complete set of methylation related genes. The D. noxia genome displays a widespread, extensive reduction in the number of genes per ortholog group, including defensive, detoxification, chemosensory, and sugar transporter groups in comparison to the Acyrthosiphon pisum genome, including a 65% reduction in chemoreceptor genes. Thirty of 34 known D. noxia salivary genes were found in this assembly. These genes exhibited less homology with those salivary genes commonly expressed in insect saliva, such as glucose dehydrogenase and trehalase, yet greater conservation among genes that are expressed in D. noxia saliva but not detected in the saliva of other insects. Genes involved in insecticide activity and endosymbiont-derived genes were also found, as well as genes involved in virus transmission, although D. noxia is not a viral vector. Conclusions This genome is the second sequenced aphid genome, and the first of a phytotoxic insect. D. noxia’s reduced gene content of may reflect the influence of phytotoxic feeding in shaping the D. noxia genome, and in turn in broadening its host range. The presence of methylation-related genes, including cytosine methylation, is consistent with other parthenogenetic and polyphenic insects. The D. noxia genome will provide an important contrast to the A. pisum genome and advance functional and comparative genomics of insects and other organisms. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1525-1) contains supplementary material, which is available to authorized users.
- Published
- 2015
35. Modification of Occupational Exposures on Bladder Cancer Risk by Common Genetic Polymorphisms
- Author
-
Ludmila Prokunina-Olsson, Stella Koutros, Molly Schwenn, Joseph F. Fraumeni, Manolis Kogevinas, Melissa C. Friesen, Stephen J. Chanock, Patricia A. Stewart, Kris Ylaya, Montserrat Garcia-Closas, Reina García-Closas, Margaret R. Karagas, Josep Lloreta, Alison Johnson, Joon-Yong Chung, Debra T. Silverman, F.X. Real, Núria Malats, A. Rouf Banday, Stephen M. Hewitt, Karla R. Armenti, Dalsu Baris, Ashley Paquin, Michael L. Nickerson, Nathaniel Rothman, Jonine D. Figueroa, Consol Serra, Adonina Tardón, Petra Lenz, Alan R. Schned, Lee E. Moore, Joanne S. Colt, Alfredo Carrato, and Nilanjan Chatterjee
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Ubiquitin-Protein Ligases ,Biology ,medicine.disease_cause ,Brief Communication ,Polymorphism, Single Nucleotide ,Germline mutation ,Risk Factors ,Internal medicine ,Occupational Exposure ,Surveys and Questionnaires ,Genetic variation ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Genetic Predisposition to Disease ,Gene–environment interaction ,Glucuronosyltransferase ,Germ-Line Mutation ,Tumor marker ,Aged ,Glutathione Transferase ,Bladder cancer ,Urinary bladder ,Environmental exposure ,Middle Aged ,medicine.disease ,Occupational Diseases ,medicine.anatomical_structure ,Scotland ,Urinary Bladder Neoplasms ,Immunology ,Metallurgy ,Female ,Gene-Environment Interaction ,Carcinogenesis ,Microtubule-Associated Proteins ,Gene Deletion - Abstract
Few studies have demonstrated gene/environment interactions in cancer research. Using data on high-risk occupations for 2258 case patients and 2410 control patients from two bladder cancer studies, we observed that three of 16 known or candidate bladder cancer susceptibility variants displayed statistically significant and consistent evidence of additive interactions; specifically, the GSTM1 deletion polymorphism ( interaction ≤ .001), rs11892031 ( UGT1A, P interaction = .01), and rs798766 (TMEM129-TACC3-FGFR3, P interaction = .03). There was limited evidence for multiplicative interactions. When we examined detailed data on a prevalent occupational exposure associated with increased bladder cancer risk, straight metalworking fluids, we also observed statistically significant additive interaction for rs798766 ( TMEM129-TACC3-FGFR3, P interaction = .02), with the interaction more apparent in patients with tumors positive for FGFR3 expression. All statistical tests were two-sided. The interaction we observed for rs798766 ( TMEM129-TACC3-FGFR3) with specific exposure to straight metalworking fluids illustrates the value of integrating germline genetic variation, environmental exposures, and tumor marker data to provide insight into the mechanisms of bladder carcinogenesis.
- Published
- 2015
36. High Frequency of Somatic Frameshift BHD Gene Mutations in Birt-Hogg-Dubé–Associated Renal Tumors
- Author
-
Youfeng Yang, Laura S. Schmidt, W. Marston Linehan, Christian P. Pavlovich, Carlos Torres-Cabala, Michael L. Nickerson, Berton Zbar, Cathy D. Vocke, Maria J. Merino, and McClellan M. Walther
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Loss of Heterozygosity ,Gene mutation ,Biology ,medicine.disease_cause ,Birt–Hogg–Dubé syndrome ,Frameshift mutation ,Loss of heterozygosity ,Germline mutation ,Gene Frequency ,Proto-Oncogene Proteins ,medicine ,Humans ,Folliculin ,Frameshift Mutation ,Tumor Suppressor Proteins ,Genodermatosis ,Proteins ,Sequence Analysis, DNA ,medicine.disease ,Kidney Neoplasms ,Oncology ,Cancer research ,Carcinogenesis - Abstract
The Birt-Hogg-Dubé (BHD) syndrome is an inherited genodermatosis characterized by a predisposition to hamartomatous skin lesions, pulmonary cysts, and renal carcinoma. Seventy-seven renal tumors from 12 patients with germline BHD mutations were examined by DNA sequencing to identify somatic mutations in the second copy of BHD. Sequence alterations were detected in the majority of renal tumors (41 of 77, 53%), with loss of heterozygosity at the BHD locus in a minority of additional tumors (14 of 77, 17%). The somatic mutations were distributed across the entire gene, and the majority resulted in frameshifts that are predicted to truncate the BHD protein. These results support a role for BHD as a tumor suppressor gene that predisposes to the development of renal tumors when both copies are inactivated.
- Published
- 2005
37. Germline BHD-Mutation Spectrum and Phenotype Analysis of a Large Cohort of Families with Birt-Hogg-Dubé Syndrome
- Author
-
Michelle B. Warren, Berton Zbar, Nirmala Sharma, Maria L. Turner, Eamonn R. Maher, W. Marston Linehan, Jorge R. Toro, Patrick J. Morrison, Maria J. Merino, McClellan M. Walther, Peter L. Choyke, Laura S. Schmidt, Gladys Glenn, James Peterson, and Michael L. Nickerson
- Subjects
Male ,Heterozygote ,Fibrofolliculoma ,Biology ,Birt–Hogg–Dubé syndrome ,Germline ,Nuclear Family ,Cohort Studies ,Exon ,Germline mutation ,Gene Frequency ,Proto-Oncogene Proteins ,Genetics ,medicine ,Adenoma, Oxyphilic ,Humans ,Genetics(clinical) ,Genetic Testing ,Folliculin ,Germ-Line Mutation ,Genetics (clinical) ,Retrospective Studies ,Patient Selection ,Tumor Suppressor Proteins ,Haplotype ,Genodermatosis ,Proteins ,Exons ,Sequence Analysis, DNA ,Syndrome ,Articles ,medicine.disease ,Introns ,Kidney Neoplasms ,Pedigree ,Phenotype ,Haplotypes ,Female - Abstract
Birt-Hogg-Dubé syndrome (BHD), a genodermatosis characterized by multiple hamartomas of the hair follicle (fibrofolliculoma), predisposes individuals to an increased risk of developing renal neoplasms and spontaneous pneumothorax. Previously, we localized the BHD locus (also known as FLCN) to chromosome 17p11.2 by linkage analysis and subsequently identified germline mutations in a novel gene in probands from eight of the nine families with BHD in our screening panel. Affected members of five of the families inherited an insertion/deletion of a cytosine in a C8 tract in exon 11. This mutation was also identified by exon 11 screening in probands from 22 of 52 additional families with BHD and therefore represents a hypermutable “hotspot” for mutation in BHD. Here, we screened the remaining 30 families from this large BHD cohort by direct sequence analysis and identified germline BHD mutations in 84% (51/61) of all families with BHD recruited to our study. Mutations were located along the entire length of the coding region, including 16 insertion/deletion, 3 nonsense, and 3 splice-site mutations. The majority of BHD mutations were predicted to truncate the BHD protein, folliculin. Among patients with a mutation in the exon 11 hotspot, significantly fewer renal tumors were observed in patients with the C-deletion than those with the C-insertion mutation. Coding-sequence mutations were not found, however, in probands from two large families with BHD whose affected members shared their family’s BHD-affected haplotype. Of the 53 families with BHD whose members inherited either a germline mutation or the affected haplotype, 24 (45%) had at least one member with renal neoplasms. Three families classified with familial renal oncocytoma were identified with BHD mutations, which represents the first disease gene associated with this rare form of renal neoplasm. This study expands the BHD-mutation spectrum and evaluates genotype-phenotype correlations among families with BHD.
- Published
- 2005
38. The Ter mutation in the dead end gene causes germ cell loss and testicular germ cell tumours
- Author
-
Xiaoning Peng, Jian Min Deng, Michael L. Nickerson, Chitralekha Bhattacharya, Blanche Capel, Joseph H. Nadeau, Laura S. Schmidt, Angabin Matin, Edward M. Rubin, Douglas Coveney, Richard R. Behringer, Kirsten K. Youngren, and Bruce T. Lamb
- Subjects
Male ,Positional cloning ,Transgene ,Testicular Germ Cell Tumor ,Gene mutation ,Biology ,medicine.disease_cause ,Article ,Embryonal carcinoma ,Mice ,Testicular Neoplasms ,Testis ,medicine ,Animals ,RNA, Messenger ,Gene ,Alleles ,In Situ Hybridization ,Genetics ,Mutation ,Multidisciplinary ,Base Sequence ,Gene Expression Profiling ,Body Weight ,Genetic Complementation Test ,Chromosome Mapping ,food and beverages ,Organ Size ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,Chromosomes, Mammalian ,Neoplasm Proteins ,Cell biology ,Disease Models, Animal ,Germ Cells ,medicine.anatomical_structure ,human activities ,Germ cell - Abstract
The phenotype of Ter testicular germ cell tumour susceptibility gene was first described more than 30 years ago, but it has taken until now for the identity of the gene to be discovered. Ter is a mutation inducing a termination codon on the mouse version of the dead end gene, known from zebrafish embryos. It encodes a protein with an RNA recognition motif, thus implicating RNA biology in testicular tumour development. In mice, the Ter mutation causes primordial germ cell (PGC) loss in all genetic backgrounds1. Ter is also a potent modifier of spontaneous testicular germ cell tumour (TGCT) susceptibility in the 129 family of inbred strains, and markedly increases TGCT incidence in 129-Ter/Ter males2,3,4. In 129-Ter/Ter mice, some of the remaining PGCs transform into undifferentiated pluripotent embryonal carcinoma cells2,3,4,5,6, and after birth differentiate into various cells and tissues that compose TGCTs. Here, we report the positional cloning of Ter, revealing a point mutation that introduces a termination codon in the mouse orthologue (Dnd1) of the zebrafish dead end (dnd) gene. PGC deficiency is corrected both with bacterial artificial chromosomes that contain Dnd1 and with a Dnd1-encoding transgene. Dnd1 is expressed in fetal gonads during the critical period when TGCTs originate. DND1 has an RNA recognition motif and is most similar to the apobec complementation factor, a component of the cytidine to uridine RNA-editing complex. These results suggest that Ter may adversely affect essential aspects of RNA biology during PGC development. DND1 is the first protein known to have an RNA recognition motif directly implicated as a heritable cause of spontaneous tumorigenesis. TGCT development in the 129-Ter mouse strain models paediatric TGCT in humans. This work will have important implications for our understanding of the genetic control of TGCT pathogenesis and PGC biology.
- Published
- 2005
39. Characterization of an S-locus receptor protein kinase-like gene from peach
- Author
-
Michael L. Nickerson, Ahmed El Ghaouth, Robert E. Farrell, Charles L. Wilson, Carole L. Bassett, Michael Wisniewski, and Timothy S. Artlip
- Subjects
Physiology ,Sequence analysis ,Acclimatization ,Molecular Sequence Data ,Plant Science ,Biology ,Prunus ,chemistry.chemical_compound ,Amino Acid Sequence ,Protein kinase A ,Gene ,Gene Library ,Plant Proteins ,Messenger RNA ,Sequence Homology, Amino Acid ,cDNA library ,Kinase ,Biological Evolution ,Molecular biology ,Cold Temperature ,chemistry ,Multigene Family ,Plant Bark ,Protein Kinases ,Salicylic acid - Abstract
A receptor-like protein kinase gene (Ppsrkl1) was isolated from a peach (Prunus persica (L.) Batsch.) bark cDNA library prepared with RNAs isolated from bark collected in December (cold acclimated). Sequence analysis indicated that this gene is related to the S-locus family of receptor protein kinases (SRKs) and that it shares greatest homology with ZMPK1 from maize and At4g32300 from Arabidopsis, both of which are intron-less genes. In bark tissues, Ppsrkl1 is induced by water deficit treatment, repressed by short-day photoperiods and showed no response to cold treatment. The Ppsrkl1 mRNA also increased in roots in response to water deficit. In fruit, Ppsrkl1 shows no response up to 6 h after wounding, but at 12 and 24 h after wounding, Ppsrkl1 mRNA shows an abrupt decline. This decline was prevented by the addition of salicylic acid to the wound site. The Ppsrkl1 mRNA rapidly decreased in fruit after 10-min exposure to UV-C radiation, followed by a return to normal levels within 1.5 h. Taken together, these experiments indicate that Ppsrkl1 is negatively regulated by light and positively influenced by salicylic acid treatment in fruit and water stress in bark and roots.
- Published
- 2005
40. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé syndrome
- Author
-
Jorge R. Toro, W. Marston Linehan, Cheryl R. Greenberg, Maria J. Merino, Nirmala Sharma, Gladys Glenn, Christian P. Pavlovich, Peter L. Choyke, David J. Munroe, McClellan M. Walther, Michelle B. Warren, Maria L. Turner, Michael L. Nickerson, Berton Zbar, Laura S. Schmidt, Paul H. Duray, Michael I. Lerman, Eamonn R. Maher, Robert Hill, and Vera Y. Matrosova
- Subjects
Male ,Cancer Research ,Candidate gene ,Estrone ,Hamartoma ,DNA Mutational Analysis ,Molecular Sequence Data ,Biology ,Birt–Hogg–Dubé syndrome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Amino Acid Sequence ,RNA, Messenger ,Folliculin ,Frameshift Mutation ,Renal oncocytoma ,Conserved Sequence ,030304 developmental biology ,Genetics ,0303 health sciences ,Base Sequence ,Genodermatosis ,Pneumothorax ,Cancer ,Exons ,Syndrome ,Cell Biology ,Physical Chromosome Mapping ,medicine.disease ,Hair follicle ,Kidney Neoplasms ,Pedigree ,3. Good health ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Hair Follicle ,Kidney cancer ,Chromosomes, Human, Pair 17 - Abstract
Birt-Hogg-Dube (BHD) syndrome is a rare inherited genodermatosis characterized by hair follicle hamartomas, kidney tumors, and spontaneous pneumothorax. Recombination mapping in BHD families delineated the susceptibility locus to 700 kb on chromosome 17p11.2. Protein-truncating mutations were identified in a novel candidate gene in a panel of BHD families, with a 44% frequency of insertion/deletion mutations within a hypermutable C 8 tract. Tissue expression of the 3.8 kb transcript was widespread, including kidney, lung, and skin. The full-length BHD sequence predicted a novel protein, folliculin, that was highly conserved across species. Discovery of disease-causing mutations in BHD , a novel kidney cancer gene associated with renal oncocytoma or chromophobe renal cancer, will contribute to understanding the role of folliculin in pathways common to skin, lung, and kidney development.
- Published
- 2002
41. Abstract 4384: Targeted sequencing reveals distinct and rare pathogenic variants in Caucasians with colorectal cancer
- Author
-
Hamed Azimi, Sudhir Varma, Hassan Ashktorab, Ali reza Safarpour, Hasti Olumi, Michael L. Nickerson, Hassan Brim, and Pooneh Mokarram
- Subjects
Genetics ,Cancer Research ,Oncology ,Colorectal cancer ,medicine ,Biology ,medicine.disease - Abstract
PURPOSE: Next-generation sequencing (NGS) is currently used to establish mutational profiles in many multigene diseases such as colorectal cancer (CRC) which is on the rise in many parts of the developing World including in the Middle East. Little is known about its genetic hallmarks in these populations. AIM: To identify variants in 15 CRC-associated genes in patients of Iranian descent. METHODS: CRC specimens from 63 patients were used to establish the variants’ profile on an Ion Torrent platform by targeted exome sequencing. To rule out technical artifacts, the variants were validated in 13 of these samples using an Illumina NGS platform. Validated variants were annotated and compared to variants from publically available databases. An in-silico functional analysis was performed. MSI status of the analyzed samples was established. RESULTS: There were 51 validated variants distributed on 12 genes: 22% MSH3 (n=11/51), 10% MSH6 (n=5/51), 8% AMER1 (n=4/51), 20% APC (n=10/51), 2% BRAF (n=1/51), 2% KRAS (n=1/51), 12% PIK3CA (n=6/51), 8% TGFβR2A (n=4/51), 2% SMAD4 (n=1/51), 4% SOX9 (n=2/51), 6% TCF7L2 (n=3/51), and 6% TP53 (n=3/51). Most known and distinct variants were in mismatch repair genes (MMR, 32%) and APC (20%). Among oncogenes, PIK3CA was the top target (12%). MSH3 variants were more frequent and predominantly homozygous in the analyzed population. CONCLUSION: These results illustrate for the first time CRC mutational profile in Iranian patients. MSH3, MSH6, APC and PIK3CA genes seem to play a bigger role in the path to cancer in this population. This is especially true for MSH3 variants that were very frequent and predominantly homozygous as these will associate with the EMAST phenotype that has prognostic implications. These findings will potentially lead to informed genetic diagnosis protocol and targeted therapeutic strategies. Citation Format: Pooneh Mokarram, Sudhir Varma, Hamed Azimi, Hasti Olumi, Ali reza Safarpour, Michael Nickerson, Hassan Brim, Hassan Ashktorab. Targeted sequencing reveals distinct and rare pathogenic variants in Caucasians with colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4384. doi:10.1158/1538-7445.AM2017-4384
- Published
- 2017
42. Abstract 3352: Genome-wide enhancer identify signature predictive of metastatic phenotypes in bladder cancers
- Author
-
Lyuba Varticovski, Michael L. Nickerson, Thompson Bethtrice, Piyush K. Agarwal, Qizong Lao, Songjoon Baek, Dan Theodorescu, Lars Grøntved, Sohyoung Kim, Michael Dean, Gordon L. Hager, and Myong-Hee Sung
- Subjects
Cancer Research ,Oncology ,Computational biology ,Biology ,Bioinformatics ,Enhancer ,Genome ,Phenotype ,Signature (logic) - Abstract
In urothelial bladder cancer accurate identification of grade and stage is critical for optimal treatment to achieve robust disease control and long-term survival. However, among the initially superficial tumors, which are non-invasive and highly treatable, 20 to 25 % recur, progress to invasive tumors, and metastasize during the patients’ lifetime. Thus, the challenge is to provide risk stratification during the initial diagnosis in order to identify those patients who are unlikely to progress while offering radical therapy to those who are at risk. To address this issue, the field has heavily focused on the discovery of few mutations in potential driver genes. However, recent findings indicate that deregulation of enhancers can play a major role in cancer progression. In this study, we employed DNase-Seq to detect enhancer activities genome-wide in 16 bladder cancer cell lines (BLCs) that included three lineages (T24, UMUC3, 253J lineages) with different tumorigenic and metastatic potentials, and thus represent models of cancer progression and metastasis development. We analyzed the gain and loss of enhancer activity in each BLC lineages as well as in metastatic cell lines relative to non-metastatic cell lines. Our analysis in the T24 lineage revealed a striking feature, a dramatic loss of intronic and distal DHSs (DNase I Hypersensitive sites), during the initial transition to tumorigenic type. During metastatic progression, new enhancer classes emerged and there was an enrichment for association with target organ-specific genes. The analysis of differential DHSs between metastatic and non-metastatic BLCs identified an enhancer signature enriched with lost DHSs in metastatic BLCs where nearby genes were associated with cellular movement/invasion functions. Lost DHSs were also present near key factors such as PPARG, RXRA, FOXA1, TP63, and GATA3, which play a role in urothelial development and differentiation, and the transcription activity of those genes were correlated with the change of enhancer activity. This study identified a set of DHSs that are associated with cancer progression in bladder cancer, and provides a potential clinical application for developing prognostic markers to predict the risk of developing aggressive disease. Citation Format: Sohyoung Kim, Lyuba Varticovski, Qizong Lao, Songjoon Baek, Michael L. Nickerson, Myong-Hee Sung, Lars Grontved, Thompson Bethtrice, Dan Theodorescu, Piyush K. Agarwal, Michael Dean, Gordon Hager. Genome-wide enhancer identify signature predictive of metastatic phenotypes in bladder cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3352. doi:10.1158/1538-7445.AM2017-3352
- Published
- 2017
43. Abstract 2379: Nuclear lemur tyrosine kinase-2 regulates RNA polymerase II dependent transcription in prostate cancer
- Author
-
Justin Foley, Neil A. Bradbury, Kalpit Shah, Michael L. Nickerson, and Michael Dean
- Subjects
Cancer Research ,Messenger RNA ,biology ,General transcription factor ,RNA polymerase II ,Molecular biology ,LMTK2 ,Cell biology ,Oncology ,Transcription (biology) ,Gene expression ,biology.protein ,Transcription factor II D ,Transcription factor - Abstract
The Androgen Receptor (AR), a DNA-binding transcription factor plays a key role in the development and maintenance of prostate epithelia by modulating expression of growth-promoting genes. AR has been proposed to regulate gene expression by enhancing the efficiency of RNA polymerase II (RNAPII) dependent transcription elongation. Dysregulation of this AR-dependent transcriptional activity has been implicated not only in prostate cancer but also androgen-independent prostate cancer. Recently, we showed that protein expression of Lemur Tyrosine Kianse-2 (LMTK2), a serine-threonine kinase is downregulated in prostate cancer. Importantly, our study ascribed a novel role for LMTK2, as a negative regulator of AR-dependent transcriptional activity in prostate epithelial cells. However, the mechanism through which LMTK2 is able to regulate AR remains to be determined. Here, we show that LMTK2 is not only present in cytoplasmic fraction but also, within the nucleus of mammalian cells where it co-localizes with AR, newly transcribed mRNA and with RNA polymerase II (RNAPII). Interestingly, our data reveals that LMTK2 colocalizes with elongating RNAPII, phosphorylated on serine 2 of the carboxyl-terminal domain. This presents an interesting possibility that LMTK2 might be able to regulate transcription of AR-dependent genes by interacting with the elongating RNAPII and transcription & epigenetic factors. As such, our study potentially identifies LMTK2 as an important player regulating the AR-RNAPII mediated transcription machinery in mammalian cells. Citation Format: Kalpit Shah, Justin Foley, Michael L. Nickerson, Michael Dean, Neil Bradbury. Nuclear lemur tyrosine kinase-2 regulates RNA polymerase II dependent transcription in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2379. doi:10.1158/1538-7445.AM2017-2379
- Published
- 2017
44. Abstract 355: TET2-loss modifies androgen signaling in prostate cancer
- Author
-
Sonja I. Berndt, Michael L. Nickerson, Kate Im, Thorkell Andresson, Joseph Boland, Jean-Noel Billaud, Hongchuan Li, Tongwu Zhang, Meredith Yeager, Stephen K. Anderson, Sevilay Turan, Seth A. Brodie, Sudipto Das, Hong Lou, Kalpit Shah, and Michael Dean
- Subjects
Cancer Research ,Prostate cancer ,Oncology ,business.industry ,medicine.drug_class ,Cancer research ,Medicine ,business ,Androgen ,medicine.disease - Abstract
Molecular markers for metastatic prostate cancer (PCa) can be identified by sequencing metastatic tumor genomes. We recently identified cancer gene mutations in a patient with metastatic disease using next generation sequencing (NGS) of DNA from 5 metastatic tumors and blood. We characterized a somatic non-conservative substitution in the methylcytosine dioxygenase ten-eleven translocation 2 (TET2) in all metastatic tumors but not the blood or primary adenocarcinoma. Analysis of metastatic tumors from additional patients revealed frequent somatic loss and sequence alteration of TET2, which was previously observed to be altered in 5-15% of myeloid, kidney and colon cancer. Genome-wide association studies previously identified non-coding risk variants associated with PCa and melanoma, and rare germline missense variants are observed in African-Americans with PCa. We sought to further investigate the role of TET2 in PCa. We performed fine-mapping of PCa risk across the TET2 locus using genotypes from the PEGASUS case-control cohort and identify six new risk variants in introns 1 and 2. Electrophoretic mobility shift assays show two risk SNPs are bound by the transcription factor octamer-binding protein 1 (Oct1). Full length TET2 (2002 aa) is expressed in normal prostate and cancer tissue and is significantly reduced in a subset of the Cancer Genome Atlas (TCGA) PCa tumors that are associated with metastatic disease and reduced disease-free survival. TET2-loss drives a cancer phenotype as siRNA-mediated knockdown (KD) significantly increases LNCaP and DU145 cell proliferation, and LNCaP transwell migration and wound healing. Affinity chromatography, mass spectrometry and co-immunoprecipitation confirm that endogenous TET2 binds the androgen receptor (AR) in LNCaP cell extracts. TET2 KD alters the expression of a subset of androgen-responsive genes including increased prostate-specific antigen (KLK3/PSA) expression, and published data indicate TET-catalyzed hydroxymethylcytosine (hmC) and TET2 binding sites proximal to KLK3. Analysis of TCGA PCa tumor RNA-seq reveals TET2 expression is co-regulated with the expression of genes encoding functions metabolizing 2-oxoglutarate and succinate including the lysine demethylase KDM6A, BRCA1-associated BAP1, and citric acid cycle enzymes IDH1-2, SDHA-B and FH. Co-expression is conserved across all 31 TCGA cancers examined. Examination of genomic locations associated with TET2-binding and hmC, and gene expression changes during androgen signaling indicate a putative role for TET2 as an energy sensor that modifies androgen-AR signaling. Decreased TET2 mRNA expression in TCGA PCa tumors that are associated with reduced patient survival indicates TET2 expression may be an informative biomarker of PCa disease progression. Citation Format: Michael L. Nickerson, Sudipto Das, Kate Im, Sevilay Turan, Sonja Berndt, Hongchuan Li, Hong Lou, Seth Brodie, Kalpit Shah, Jean-Noel Billaud, Tongwu Zhang, Joseph Boland, Stephen Anderson, Meredith Yeager, Michael Dean, Thorkell Andresson. TET2-loss modifies androgen signaling in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 355. doi:10.1158/1538-7445.AM2017-355
- Published
- 2017
45. Distinctive DNA Mismatch Repair and APC Variants in African American with Colorectal Neoplasia
- Author
-
Michael L. Nickerson, Sudhir Varma, Hassan Ashktorab, Hamed Azimi, and Hassan Brim
- Subjects
African american ,Hepatology ,Gastroenterology ,Cancer research ,DNA mismatch repair ,Biology - Published
- 2017
46. Signature-based analysis of MET proto-oncogene mutations using DHPLC
- Author
-
Berton Zbar, Michael L. Nickerson, Gregor Weirich, and Laura S. Schmidt
- Subjects
Genetics ,DNA profiling ,Mutant ,DNA Mutational Analysis ,Wild type ,SNP ,Single-nucleotide polymorphism ,Biology ,Allele ,Genetics (clinical) ,Heteroduplex - Abstract
Research tools which improve mutation detection, SNP discovery, and allele characterization will facilitate studies of cancer, inherited disease, and genomic evolution. Denaturing High-Performance Liquid Chromatography (DHPLC) is a recently developed methodology for detection of heteroduplexes formed in DNA samples containing mismatches between wild type and mutant strands. In an effort to develop a rapid, sensitive mutation detection method for studies of families with inherited kidney cancer, we evaluated DHPLC for detection and analysis of MET proto-oncogene mutations in papillary renal carcinomas (PRC). We found DHPLC to be 100% accurate in detecting 15 known disease-associated MET mutations. Significantly, each MET mutation and two novel SNPs generated a characteristic chromatographic profile or signature with reproducible distinguishing features. Standardization of DHPLC reagents and improved methods design were critical to the reliability and accuracy of mutation prediction. Improvements included addition of a 75% acetonitrile wash followed by a rejuvenating gradient, and detailed analysis of signature shape, retention time (RT), RT differences (DeltaRT), and temperature-dependent (melt) profiling. We used signatures to predict mutations in new PRC samples, mutation carriers in asymptomatic hereditary PRC family members, and in a blind study of previously characterized DNAs. Application to SNP discovery is discussed. Wiley-Liss, Inc.
- Published
- 2000
47. [Untitled]
- Author
-
Reuben A. Cohen, Mangalathu S. Rajeevan, Michael L. Nickerson, and Carole L. Bassett
- Subjects
Messenger RNA ,Alternative splicing ,Intron ,Plant Science ,General Medicine ,Leucine-rich repeat ,Biology ,Molecular biology ,Post-transcriptional modification ,Transcription (biology) ,Gene expression ,Genetics ,Agronomy and Crop Science ,Gene - Abstract
A gene (inrpk1) encoding a putative receptor-like protein kinase was isolated from the Japanese morning glory, Ipomoea (Pharbitis) nil Roth. cv. Violet. The receptor-like portion of the largest derived polypeptide contains 26 direct leucine-rich repeats (LRRs) in a single block, and the catalytic portion has all the conserved amino acid residues characteristic of Ser/Thr protein kinases. RNA blot analysis detected multiple transcripts in cotyledons. The largest (4.4 kb) transcript encodes the predicted full length polypeptide (INRPK1), whereas a 1.6 kb transcript apparently originates from a secondary transcription initiation site within the gene and potentially encodes a protein kinase identical to INRPK1, but lacking most of the LRRs. Two transcripts (ca. 2.7 and 2.6 kb) are created by alternative 3′-splicing of a large (ca. 1.4–1.5 kb) cryptic intron in the LRR region, creating one transcript (2.6 kb) potentially encoding a small, secretable polypeptide. The larger transcript encoding a polypeptide identical to INRPK1, but lacking 21 LRRs, predominates in vegetative roots. Competitive PCR indicates that inrpk1 mRNA increases 20-fold in cotyledons in response to a previously given single floral-inducing short-day (SD). No differences of this magnitude were detected in any other organs examined from plants similarly treated. This pattern of expression and differential processing suggests a role for inrpk1 in some aspect of SD photoperiodic-induced flowering in morning glory.
- Published
- 2000
48. The TERE1 protein interacts with mitochondrial TBL2: regulation of trans-membrane potential, ROS/RNS and SXR target genes
- Author
-
William J, Fredericks, Terry, McGarvey, Huiyi, Wang, Yongmu, Zheng, Nathaniel J, Fredericks, Hankun, Yin, Li-Ping, Wang, Wayland, Hsiao, Rob, Lee, Jayne S, Weiss, Michael L, Nickerson, Howard S, Kruth, Frank J, Rauscher, and S Bruce, Malkowicz
- Subjects
Reverse Transcriptase Polymerase Chain Reaction ,Dimethylallyltranstransferase ,Lipid Metabolism ,Reactive Nitrogen Species ,Cell Line ,Membrane Potentials ,Mitochondria ,Oxidative Stress ,GTP-Binding Proteins ,Humans ,Immunoprecipitation ,Fluorescent Antibody Technique, Indirect ,Microscopy, Immunoelectron ,Reactive Oxygen Species ,Protein Binding - Abstract
We originally discovered TERE1 as a potential tumor suppressor protein based upon reduced expression in bladder and prostate cancer specimens and growth inhibition of tumor cell lines/xenografts upon ectopic expression. Analysis of TERE1 (aka UBIAD1) has shown it is a prenyltransferase enzyme in the natural bio-synthetic pathways for both vitamin K-2 and COQ10 production and exhibits multiple subcellular localizations including mitochondria, endoplasmic reticulum, and golgi. Vitamin K-2 is involved in mitochondrial electron transport, SXR nuclear hormone receptor signaling and redox cycling: together these functions may form the basis for tumor suppressor function. To gain further insight into mechanisms of growth suppression and enzymatic regulation of TERE1 we isolated TERE1 associated proteins and identified the WD40 repeat, mitochondrial protein TBL2. We examined whether disease specific mutations in TERE1 affected interactions with TBL2 and the role of each protein in altering mitochondrial function, ROS/RNS production and SXR target gene regulation. Biochemical binding assays demonstrated a direct, high affinity interaction between TERE1 and TBL2 proteins; TERE1 was localized to both mitochondrial and non-mitochondrial membranes whereas TBL2 was predominantly mitochondrial; multiple independent single amino acid substitutions in TERE1 which cause a human hereditary corneal disease reduced binding to TBL2 strongly suggesting the relevance of this interaction. Ectopic TERE1 expression elevated mitochondrial trans-membrane potential, oxidative stress, NO production, and activated SXR targets. A TERE1-TBL2 complex likely functions in oxidative/nitrosative stress, lipid metabolism, and SXR signaling pathways in its role as a tumor suppressor.
- Published
- 2013
49. Somatic alterations contributing to metastasis of a castration-resistant prostate cancer
- Author
-
Héctor Corrada Bravo, W. Marston Linehan, Timothy T. Harkins, Berton Zbar, G. Steven Bova, Kate M. Im, Kevin J. Misner, Hong Lou, Bert Gold, Karin M. Fredrikson, Michael L. Nickerson, Meredith Yeager, Wei Tan, David Wells, Michael Dean, Thorkell Andresson, and Patrice M. Milos
- Subjects
Male ,Oncogene Proteins, Fusion ,Somatic cell ,Molecular Sequence Data ,Genes, BRCA1 ,Mutation, Missense ,Loss of Heterozygosity ,Biology ,Polymorphism, Single Nucleotide ,Germline ,Article ,Frameshift mutation ,Metastasis ,Dioxygenases ,Loss of heterozygosity ,Prostate cancer ,Germline mutation ,Proto-Oncogene Proteins ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,Neoplasm Metastasis ,Frameshift Mutation ,Genetics (clinical) ,Germ-Line Mutation ,Phylogeny ,Aged ,Nuclear Proteins ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Primary tumor ,DNA-Binding Proteins ,Prostatic Neoplasms, Castration-Resistant ,Cancer research ,Transcription Factors - Abstract
Metastatic castration resistant prostate cancer (mCRPC) is a lethal disease and molecular markers that differentiate indolent from aggressive subtypes are needed. We sequenced the exomes of five metastatic tumors and healthy kidney tissue from an index case with mCRPC to identify lesions associated with disease progression and metastasis. An Ashkenazi Jewish (AJ) germline founder mutation, del185AG in BRCA1, was observed and AJ ancestry was confirmed. Sixty-two somatic variants altered proteins in tumors, including cancer-associated genes, TMPRSS2-ERG, PBRM1, and TET2. The majority (n=53) of somatic variants were present in all metastases and only a subset (n=31) was observed in the primary tumor. Integrating tumor next generation sequencing (NGS) and DNA copy number showed somatic loss of BRCA1 and TMPRSS2-ERG. We sequenced 19 genes with deleterious mutations in the index case in additional mCRPC samples and detected a frameshift, two somatic missense alterations, tumor loss of heterozygosity (LOH), and combinations of germline missense SNPs in TET2. In summary, genetic analysis of metastases from an index case permitted us to infer a chronology for the clonal spread of disease based on sequential accrual of somatic lesions. The role of TET2 in mCRPC deserves additional analysis and may define a subset of metastatic disease.
- Published
- 2013
50. Genomic copy number alterations in clear cell renal carcinoma: associations with case characteristics and mechanisms of VHL gene inactivation
- Author
-
Ruth M. Pfeiffer, Paul Brennan, Vladimir Bencko, Maria J. Merino, Stephen M. Hewitt, Sara Karami, F. M. Waldman, Vladimir Janout, Nathanial Rothman, Erich Jaeger, Lee E. Moore, Jeffry P. Simko, Petra Lenz, H. Li, David Zaridze, Ritu Roy, Wong-Ho Chow, Michael L. Nickerson, Dana Mates, Neonilia Szeszenia-Dabrowska, S. De Vries, Marie Navratilova, W. M. Linehan, Paolo Boffetta, Jorge R. Toro, Moore, L.E., Jaeger, E., Nickerson, M.L., Brennan, P., De Vries, S., Roy, R., Toro, J., Li, H., Karami, S., Lenz, P., Zaridze, D., Janout, V., Bencko, V., Navratilova, M., Szeszenia-Dabrowska, N., Mates, D., Linehan, W.M., Merino, M., Simko, J., Pfeiffer, R., Boffetta, P., Hewitt, S., Rothman, N., Chow, W.-H., and Waldman, F.M.
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Kidney Disease ,renal cancer ,Oncology and Carcinogenesis ,Locus (genetics) ,Rare Diseases ,Clinical Research ,FHIT ,Internal medicine ,VHL ,Epidemiology ,Genetics ,Medicine ,Molecular Biology ,Gene ,Cancer ,renal cancer, epidemiology, VHL ,business.industry ,Chromosomal fragile site ,Breakpoint ,medicine.disease ,Clear cell renal cell carcinoma ,Original Article ,epidemiology ,Biochemistry and Cell Biology ,business ,Comparative genomic hybridization - Abstract
Array comparative genomic hybridization was used to identify copy number alterations in clear cell renal cell carcinoma (ccRCC) patient tumors to identify associations with patient/clinical characteristics. Of 763 ccRCC patients, 412 (54%) provided frozen biopsies. Clones were analyzed for significant copy number differences, adjusting for multiple comparisons and covariates in multivariate analyses. Frequent alterations included losses on: 3p (92.2%), 14q (46.8%), 8p (38.1%), 4q (35.4%), 9p (32.3%), 9q (31.8%), 6q (30.8%), 3q (29.4%), 10q (25.7%), 13q (24.5%), 1p (23.5%) and gains on 5q (60.2%), 7q (39.6%), 7p (30.6%), 5p (26.5%), 20q (25.5%), 12q (24.8%), 12p (22.8%). Stage and grade were associated with 1p, 9p, 9q, 13q and 14q loss and 12q gain. Males had more alterations compared with females, independent of stage and grade. Significant differences in the number/types of alterations were observed by family cancer history, age at diagnosis and smoking status. Von Hippel-Lindau (VHL) gene inactivation was associated with 3p loss (PE-05), and these cases had fewer alterations than wild-type cases. The fragile site flanking the FHIT locus (3p14.2) represented a unique breakpoint among VHL hypermethylated cases, compared with wild-type cases and those with sequence changes. This is the first study of its size to investigate copy number alterations among cases with extensive patient, clinical/risk factor information. Patients characterized by VHL wild-type gene status (vs sequence alterations) and male (vs female) cases had more copy number alterations regardless of diagnostic stage and grade, which could relate to poor prognosis. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.