1
|
Briggs LG, Steele GL, Qian ZJ, Subbana S, Alkhatib KY, Labban M, Langbein BJ, Nguyen DD, Cellini J, Kilbridge K, Kibel AS, Trinh QD, Rana HQ, Cole AP. Racial Differences in Germline Genetic Testing for Prostate Cancer: A Systematic Review. JCO Oncol Pract 2023; 19:e784-e793. [PMID: 36649495 DOI: 10.1200/op.22.00634] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Testing for pathogenic variants can aid in oncologic risk stratification and identification of targeted therapies. Despite known disparities in access to prostate cancer (PCa) care, little has been written about access to germline genetic testing (GGT) for Black men and other historically marginalized populations. This systematic review sought to delineate racial/ethnic disparities in GGT for PCa. METHODS This systematic review identified articles published from January 1996 through May 2021 in PubMed, Web of Science, and Embase. We included studies that reported rates of GGT in men with PCa in the United States by race/ethnicity as reflective of routine clinical care or research. A narrative synthesis was performed. RESULTS Of 4,309 unique records, 91 studies examining 50 unique study populations met inclusion criteria. Of these, four populations included men who received GGT through routine clinical care, accounting for 4,415 men (72.6% White and 7.2% Black). The other 46 populations included men who received GGT as part of a research study, accounting for 30,824 men (64.3% White and 21.6% Black). Of these 46 research populations, 19 used targeted methods to increase recruitment from a specific demographic. CONCLUSION Most studies that report GGT rates by race/ethnicity are in research settings. Many of these studies used targeted recruitment methods and subsequently have a greater proportion of Black men than clinical and US population-based studies. Other historically marginalized populations are not well represented. There remains a knowledge gap regarding the extent of racial disparities in the use of GGT, particularly in the clinical setting.
Collapse
Affiliation(s)
- Logan G Briggs
- Department of Urologic Surgery, Mayo Clinic, Phoenix, AZ.,Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Grant L Steele
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Zhiyu Jason Qian
- Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | | | - Khalid Y Alkhatib
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Muhieddine Labban
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Bjoern J Langbein
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David-Dan Nguyen
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Kerry Kilbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Adam S Kibel
- Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Quoc-Dien Trinh
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Huma Q Rana
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA
| | - Alexander P Cole
- Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
2
|
Kadanga E, Zouré AA, Zohoncon TM, Traoré L, Ky BD, Yonli AT, Traoré DDA, Bazié BVJTE, Sombié HK, Sorgho PA, Tovo SFA, Traoré K, Ouedraogo TWC, Djigma FW, Simpore J. Carriage of mutations R462Q (rs 486907) and D541E (rs 627928) of the RNASEL gene and risk factors in patients with prostate cancer in Burkina Faso. BMC Med Genomics 2022; 15:123. [PMID: 35655265 PMCID: PMC9161613 DOI: 10.1186/s12920-022-01279-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Prostate cancer (Pca) is a public health problem that affects men, usually of middle age or older. It is the second most common cancer diagnosed in men and the fifth leading cause of death. The RNASEL gene located in 1q25 and identified as a susceptibility gene to hereditary prostate cancer, has never been studied in relation to prostate cancer in Burkina Faso. The aim of this study was to analyze the carriage of RNASEL R462Q and D541E mutations and risks factors in patients with prostate cancer in the Burkina Faso. Methods This case–control study included of 38 histologically diagnosed prostate cancer cases and 53 controls (cases without prostate abnormalities). Real-time PCR genotyping of R462Q and D541E variants using the TaqMan® allelic discrimination technique was used. Correlations between different genotypes and combined genotypes were investigated. Results The R462Q variant was present in 5.3% of cases and 7.5% of controls. The D541E variant was present in 50.0% of cases and 35% of controls. There is no association between R462Q variants (OR = 0.60; 95%IC, 0.10–3.51; p = 0.686) and D541E variants (OR = 2.46; 95%IC, 0.78–7.80; p = 0.121) and genotypes combined with prostate cancer. However, there is a statistically significant difference in the distribution of cases according to the PSA rate at diagnosis (p ˂ 0.001). For the Gleason score distribution, only 13.2% of cases have a Gleason score greater than 7. There is a statistically significant difference in the Gleason score distribution of cases (p ˂ 0.001). Conclusions These variants, considered in isolation or in combination, are not associated with the risk of prostate cancer.
Collapse
|
3
|
Ho WHJ, Law AMK, Masle-Farquhar E, Castillo LE, Mawson A, O'Bryan MK, Goodnow CC, Gallego-Ortega D, Oakes SR, Ormandy CJ. Activation of the viral sensor oligoadenylate synthetase 2 (Oas2) prevents pregnancy-driven mammary cancer metastases. Breast Cancer Res 2022; 24:31. [PMID: 35505346 PMCID: PMC9066770 DOI: 10.1186/s13058-022-01525-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The interferon response can influence the primary and metastatic activity of breast cancers and can interact with checkpoint immunotherapy to modulate its effects. Using N-ethyl-N-nitrosourea mutagenesis, we found a mouse with an activating mutation in oligoadenylate synthetase 2 (Oas2), a sensor of viral double stranded RNA, that resulted in an interferon response and prevented lactation in otherwise healthy mice. METHODS To determine if sole activation of Oas2 could alter the course of mammary cancer, we combined the Oas2 mutation with the MMTV-PyMT oncogene model of breast cancer and examined disease progression and the effects of checkpoint immunotherapy using Kaplan-Meier survival analysis with immunohistochemistry and flow cytometry. RESULTS Oas2 mutation prevented pregnancy from increasing metastases to lung. Checkpoint immunotherapy with antibodies against programmed death-ligand 1 was more effective when the Oas2 mutation was present. CONCLUSIONS These data establish OAS2 as a therapeutic target for agents designed to reduce metastases and increase the effectiveness of checkpoint immunotherapy.
Collapse
Affiliation(s)
- Wing-Hong Jonathan Ho
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Andrew M K Law
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Etienne Masle-Farquhar
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Lesley E Castillo
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Amanda Mawson
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Moira K O'Bryan
- The School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Christopher C Goodnow
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,Cellular Genomics Futures Institute, UNSW Sydney, Kensington, NSW, Australia
| | - David Gallego-Ortega
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia.,School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, 81 Broadway, Ultimo Sydney, NSW, 2007, Australia
| | - Samantha R Oakes
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia.,National Breast Cancer Foundation Level 7, 50 Margaret Street, Sydney, NSW, 2001, Australia
| | - Christopher J Ormandy
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst Sydney, NSW, 2010, Australia. .,St. Vincent's Clinical School, St. Vincent's Hospital, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
4
|
Sienkiewicz K, Yang C, Paschal BM, Ratan A. Genomic analyses of the metastasis-derived prostate cancer cell lines LNCaP, VCaP, and PC3-AR. Prostate 2022; 82:442-451. [PMID: 34951700 PMCID: PMC8792310 DOI: 10.1002/pros.24290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND The lymph node metastasis-derived LNCaP, the bone metastasis-derived PC3 (skull), and VCaP (vertebral) cell lines are widely used as preclinical models of human prostate cancer (CaP) and have been described in more than 19,000 publications. Here, we report on short-read whole-genome sequencing and genomic analyses of LNCaP, VCaP, and PC3 cells stably transduced with WT AR (PC3-AR). METHODS LNCaP, VCaP, and PC3-AR cell lines were sequenced to an average depth of more than 30-fold using Illumina short-read sequencing. Using various computational methods, we identified and compared the single-nucleotide variants, copy-number profiles, and the structural variants observed in the three cell lines. RESULTS LNCaP cells are composed of multiple subpopulations, which results in nonintegral copy number states and a high mutational load when the data is analyzed in bulk. All three cell lines contain pathogenic mutations and homozygous deletions in genes involved in DNA mismatch repair, along with deleterious mutations in cell-cycle, Wnt signaling, and other critical cellular processes. PC3-AR cells have a truncating mutation in TP53 and do not express the p53 protein. The VCaP cells contain a homozygous gain-of-function mutation in TP53 (p.R248W) that promotes cancer invasion, metastasis, and progression and has also been observed in prostate adenocarcinomas. In addition, we detect the signatures of chromothripsis of the q arms of chromosome 5 in both PC3-AR and VCaP cells, strengthening the association of TP53 inactivation with chromothripsis reported in other systems. CONCLUSIONS Our work provides a resource for genetic, genomic, and biological studies employing these commonly-used prostate cancer cell lines.
Collapse
Affiliation(s)
| | - Chunsong Yang
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Bryce M. Paschal
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Virginia, USA
- Department of Public Health Sciences, University of Virginia, Virginia, USA
| |
Collapse
|
5
|
Wang J, Yuan W, Yue C, Dai F, Gao S, Mi Y, Bai Y, Zhang L, Zuo L, Wu X, Zhang W. RNASEL 1623A>C variant is associated with the risk of prostate cancer in African descendants. J Cell Biochem 2019; 120:11955-11964. [PMID: 30790337 DOI: 10.1002/jcb.28479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 01/24/2023]
Abstract
Association between ribonuclease L (RNASEL) gene 1623A>C polymorphism and prostate cancer (PCa) susceptibility has been assessed in large quantities of studies but with controversial conclusions. We undertook a pooled analysis containing 7397 PCa cases and 6088 control subjects to assess the correlation between RNASEL 1623A>C polymorphism and PCa risk. Moreover, we used enzyme-linked immunosorbent assay to test the serum RNASEL expression among patients enrolled in our centers and in-silico tools were also utilized. The overall results of our analysis indicated a positive relationship between 1623A>C variant and PCa risk (allelic contrast, odds ratio [OR] = 1.07; 95% confidence interval [CI] = 1.02-1.12; Pheterogeneity = 0.575; CC vs AA, OR = 1.14; 95% CI = 1.03-1.26; Pheterogeneity = 0.217; CC + CA vs AA, OR = 1.10; 95% CI = 1.01-1.19; Pheterogeneity = 0.303; and CC vs CA + AA, OR = 1.08; 95% CI = 1.00-1.17; Pheterogeneity = 0.298). In ethnicity subgroup analysis, similar results were especially indicated in African descendants. In addition, serum RNASEL levels in PCa cases with CC + CA genotypes were higher than those with AA genotypes. Our present study showed evidence that RNASEL 1623A>C polymorphism is related to PCa risk, especially in African descendants.
Collapse
Affiliation(s)
- Jian Wang
- Department of Urology, Third Affiliated Hospital of Nantong University (Affiliated Hospital of Jiangnan University), Wuxi, China
| | - Wei Yuan
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Chuang Yue
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Feng Dai
- Department of Urology, Third Affiliated Hospital of Nantong University (Affiliated Hospital of Jiangnan University), Wuxi, China
| | - Shenglin Gao
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Third Affiliated Hospital of Nantong University (Affiliated Hospital of Jiangnan University), Wuxi, China
| | - Yu Bai
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Lifeng Zhang
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Li Zuo
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Xingyu Wu
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Wei Zhang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| |
Collapse
|
6
|
Zhang Y, Jiang F, He H, Ye J, Mao X, Guo Q, Wu SL, Zhong W, Wu CL, Lin N. Identification of a novel microRNA-mRNA regulatory biomodule in human prostate cancer. Cell Death Dis 2018; 9:301. [PMID: 29467540 PMCID: PMC5833360 DOI: 10.1038/s41419-018-0293-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/06/2017] [Accepted: 01/04/2018] [Indexed: 12/29/2022]
Abstract
Our recent study identified a list of differentially expressed microRNAs (miRNAs) in human prostate cancer (PCa) tissues compared to adjacent benign prostate tissues. In the current study, to identify the crucial miRNA-mRNA regulatory biomodule involved into prostate carcinogenesis based on the previous miRNA expression profile in PCa, we proposed an integrated systematic approach which combined miRNA-mediated gene expression regulatory network analysis, experimental validations in vitro and in vivo, as well as clinical significance evaluation. As a result, the CCND1-RNASEL-CDKN1A-TP73-MDM2-UBE2I axis was identified as a bottleneck in the miRNA-mediated gene expression regulatory network of PCa according to network topological analysis. The direct binding relationship between TP73 and PCa downregulated miR-193a-5p, and the direct binding relationship between UBE2I and PCa upregulated miR-188-5p were both experimentally validated. In addition, miR-193a-5p had a more significant regulatory effect on the tumor promoter isoform of TP73-deltaNp73 than on the tumor suppressive isoform of TP73-TAp73. Importantly, the deregulation of either the miR-193a-5p-TP73 or miR-188-5p-UBE2I axes was significantly associated with aggressive progression and poor prognosis in PCa patients. Gain- and loss-of-function experiments showed that miR-193a-5p efficiently inhibited in vitro PCa cell proliferation, migration, and invasion, and in vivo tumor growth, and markedly induced PCa cell apoptosis via regulating TP73 with a corresponding suppression of the CCND1-RNASEL-CDKN1A-MDM2 axis. In contrast, miR-188-5p exerted its tumor promoter roles through targeting UBE2I with a subsequent activation of the CCND1-RNASEL-CDKN1A-MDM2 axis. Taken together, this integrated analysis revealed the potential roles of the miR-193a-5p/TP73 and miR-188-5p/UBE2i negative regulation pairs in PCa. In addition to the significant clinical relevance, miR-193a-5p- and miR-188-5p-regulated CCND1-RNASEL-CDKN1A-TP73-MDM2-UBE2I signaling may be a novel regulatory biomodule in prostate carcinogenesis.
Collapse
Affiliation(s)
- Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.,Department of Urology & Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Funeng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Huichan He
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Jianheng Ye
- Department of Urology & Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Xia Mao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiuyan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shu-Lin Wu
- Department of Urology & Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China. .,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China.
| | - Chin-Lee Wu
- Department of Urology & Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
7
|
Abstract
RNase L is a regulated endoribonuclease that functions in the interferon antiviral response. Activation of RNase L by 2', 5'-oligoadenylates has been linked to apoptosis, autophagy and inflammation. Genetic studies have also suggested the possible involvement of the RNase L gene (RNASEL) on chromosome 1q25.3 in several types of cancer. Here we report that ablation of RNase L in human prostate cancer PC3 cells by CRISPR/Cas9 gene editing technology enhanced cell migration as determined both by transwell assays and scratch wound healing assays. In addition, RNase L knockdown by means of RNAi increased migration of PC3 and DU145 cells in response to either fibronectin or serum stimulation, as did homozygous disruption of the RNase L gene in mouse embryonic fibroblasts. Serum or fibronectin stimulation of focal adhesion kinase (FAK) autophosphorylation on tyrosine-397 was increased by either knockdown or ablation of RNase L. In contrast, a missense mutant RNase L (R667A) lacking catalytic activity failed to suppress cell migration in PC3 cells. However, a nuclease-inactive mutant mouse RNase L (W630A) was able to partially inhibit migration of mouse fibroblasts. Consistent with a role for the catalytic activity of RNase L, transfection of PC3 cells with the RNase L activator, 2', 5'-oligoadenylate, suppressed cell migration. RNase L knockdown in PC3 cells enhanced tumor growth and metastasis following implantation in the mouse prostate. Our results suggest that naturally occurring mutations in the RNase L gene might promote enhanced cell migration and metastasis.
Collapse
|
8
|
Gusho E, Baskar D, Banerjee S. New advances in our understanding of the "unique" RNase L in host pathogen interaction and immune signaling. Cytokine 2016; 133:153847. [PMID: 27595182 PMCID: PMC7128181 DOI: 10.1016/j.cyto.2016.08.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/08/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Ever since the discovery of the existence of an interferon (IFN)-regulated ribonuclease, significant advances have been made in understanding the mechanism and associated regulatory effects of its action. What had been studied initially as a "unique" endoribonuclease is currently known as ribonuclease L (RNase L where "L" stands for latent). Some of the key developments include discovery of the RNase L signaling pathway, its structural characterization, and its molecular cloning. RNase L has been implicated in antiviral and antibacterial defense, as well as in hereditary prostate cancer. RNase L is activated by 2'-5' linked oligoadenylates (2-5A), which are synthesized by the oligoadenylate synthetases (OASs), a family of IFN-regulated pathogen recognition receptors that sense double-stranded RNAs. Activated RNase L cleaves single stranded RNAs, including viral RNAs and cellular RNAs. The catalytic activity of RNase L has been found to lead into the activation of several cellular signaling pathways, including those involved in autophagy, apoptosis, IFN-β production, NLRP3 inflammasome activation leading to IL-1β secretion, inhibition of cell migration, and cell adhesion. In this review, we will highlight the newest advances in our understanding of the catalytic role of RNase L in the context of different cellular pathways and extend the scope of these findings to discussion of potential therapeutic targets for antimicrobial drug development.
Collapse
Affiliation(s)
- Elona Gusho
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue Cleveland, OH 44195, USA
| | - Danika Baskar
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue Cleveland, OH 44195, USA; Pediatrics Division Office, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA(1)
| | - Shuvojit Banerjee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue Cleveland, OH 44195, USA.
| |
Collapse
|
9
|
Winchester DA, Till C, Goodman PJ, Tangen CM, Santella RM, Johnson-Pais TL, Leach RJ, Xu J, Zheng SL, Thompson IM, Lucia MS, Lippmann SM, Parnes HL, Dluzniewski PJ, Isaacs WB, De Marzo AM, Drake CG, Platz EA. Variation in genes involved in the immune response and prostate cancer risk in the placebo arm of the Prostate Cancer Prevention Trial. Prostate 2015; 75:1403-18. [PMID: 26047319 PMCID: PMC4536102 DOI: 10.1002/pros.23021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/27/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND We previously found that inflammation in benign prostate tissue is associated with an increased odds of prostate cancer, especially higher-grade disease. Since part of this link may be due to genetics, we evaluated the association between single nucleotide polymorphisms (SNPs) in immune response genes and prostate cancer in the placebo arm of the Prostate Cancer Prevention Trial. METHODS We genotyped 16 candidate SNPs in IL1β, IL2, IL4, IL6, IL8, IL10, IL12(p40), IFNG, MSR1, RNASEL, TLR4, and TNFA and seven tagSNPs in IL10 in 881 prostate cancer cases and 848 controls negative for cancer on an end-of-study biopsy. Cases and controls were non-Hispanic white and frequency matched on age and family history. We classified cases as lower (Gleason sum <7; N = 674) and higher (7-10; N = 172) grade, and used logistic regression to estimate odds ratios (OR) and 95% confidence intervals (CI) adjusting for age and family history. RESULTS The minor allele (C) of rs3212227 in IL12(p40) was associated with an increased risk of total (log additive: OR = 1.30, 95%CI 1.10-1.53; P-trend = 0.0017) and lower-grade (OR = 1.36, 95%CI 1.15-1.62; P-trend = 0.0004) prostate cancer. The minor allele (A) of rs4073 in IL8 was possibly associated with a decreased risk of higher-grade (OR = 0.81, 95%CI 0.64-1.02; P-trend = 0.07), but not total disease. None of the other candidates was associated with risk. The minor alleles of IL10 tagSNPs rs1800890 (A; OR = 0.87, 95%CI: 0.75-0.99; P-trend = 0.04) and rs3021094 (C; OR = 1.31, 95%CI 1.03-1.66, P-trend = 0.03) were associated with risk; the latter also with lower- (P-trend = 0.04) and possibly higher- (P-trend = 0.06) grade disease. These patterns were similar among men with PSA <2 ng/ml at biopsy. CONCLUSION Variation in some immune response genes may be associated with prostate cancer risk. These associations were not fully explained by PSA-associated detection bias. Our findings generally support the role of inflammation in the etiology of prostate cancer.
Collapse
Affiliation(s)
- Danyelle A. Winchester
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Cathee Till
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis J. Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Regina M. Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Teresa L. Johnson-Pais
- Department of Urology, University of Texas Health Sciences Center San Antonio, San Antonio, TX
| | - Robin J. Leach
- Department of Urology, University of Texas Health Sciences Center San Antonio, San Antonio, TX
| | - Jianfeng Xu
- Program for Personalized Cancer Care and Department of Surgery, NorthShore University Health System, Evanston, IL
| | - S. Lilly Zheng
- Program for Personalized Cancer Care and Department of Surgery, NorthShore University Health System, Evanston, IL
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ian M. Thompson
- Department of Urology, University of Texas Health Sciences Center San Antonio, San Antonio, TX
| | - M. Scott Lucia
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO
| | - Scott M. Lippmann
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Howard L. Parnes
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Paul J. Dluzniewski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - William B. Isaacs
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Angelo M. De Marzo
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Charles G. Drake
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
- Department of Immunology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
10
|
Farzan SF, Karagas MR, Christensen BC, Li Z, Kuriger JK, Nelson HH. RNASEL and MIR146A SNP-SNP interaction as a susceptibility factor for non-melanoma skin cancer. PLoS One 2014; 9:e93602. [PMID: 24699816 PMCID: PMC3974770 DOI: 10.1371/journal.pone.0093602] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 03/06/2014] [Indexed: 02/02/2023] Open
Abstract
Immunity and inflammatory pathways are important in the genesis of non-melanoma skin cancers (NMSC). Functional genetic variation in immune modulators has the potential to affect disease etiology. We investigated associations between common variants in two key regulators, MIR146A and RNASEL, and their relation to NMSCs. Using a large population-based case-control study of basal cell (BCC) and squamous cell carcinoma (SCC), we investigated the impact of MIR146A SNP rs2910164 on cancer risk, and interaction with a SNP in one of its putative targets (RNASEL, rs486907). To examine associations between genotype and BCC and SCC, occurrence odds ratios (OR) and 95% confidence intervals (95%CI) were calculated using unconditional logistic regression, accounting for multiple confounding factors. We did not observe an overall change in the odds ratios for SCC or BCC among individuals carrying either of the RNASEL or MIR146A variants compared with those who were wild type at these loci. However, there was a sex-specific association between BCC and MIR146A in women (ORGC = 0.73, [95%CI = 0.52-1.03]; ORCC = 0.29, [95% CI = 0.14-0.61], p-trend<0.001), and a reduction in risk, albeit not statistically significant, associated with RNASEL and SCC in men (ORAG = 0.88, [95%CI = 0.65-1.19]; ORAA = 0.68, [95%CI = 0.43-1.08], p-trend = 0.10). Most striking was the strong interaction between the two genes. Among individuals carrying variant alleles of both rs2910164 and rs486907, we observed inverse relationships with SCC (ORSCC = 0.56, [95%CI = 0.38-0.81], p-interaction = 0.012) and BCC (ORBCC = 0.57, [95%CI = 0.40-0.80], p-interaction = 0.005). Our results suggest that genetic variation in immune and inflammatory regulators may influence susceptibility to NMSC, and novel SNP-SNP interaction for a microRNA and its target. These data suggest that RNASEL, an enzyme involved in RNA turnover, is controlled by miR-146a and may be important in NMSC etiology.
Collapse
Affiliation(s)
- Shohreh F. Farzan
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Margaret R. Karagas
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Brock C. Christensen
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Zhongze Li
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Jacquelyn K. Kuriger
- Division of Epidemiology and Community Health, The Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Heather H. Nelson
- Division of Epidemiology and Community Health, The Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | | |
Collapse
|
11
|
Schoenfeld JD, Margalit DN, Kasperzyk JL, Shui IM, Rider JR, Epstein MM, Meisner A, Kenfield SA, Martin NE, Nguyen PL, Kantoff PW, Giovannucci EL, Stampfer MJ, Mucci LA. A single nucleotide polymorphism in inflammatory gene RNASEL predicts outcome after radiation therapy for localized prostate cancer. Clin Cancer Res 2013; 19:1612-9. [PMID: 23382116 PMCID: PMC3602407 DOI: 10.1158/1078-0432.ccr-12-2718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE To study associations between single nucleotide polymorphisms (SNP) in Ribonuclease L (RNASEL), a gene implicated in inflammation and prostate cancer risk, and outcomes after radiation therapy. EXPERIMENTAL DESIGN We followed participants in the prospective US Health Professionals Follow-Up Study treated with radiation therapy for early-stage prostate cancer. Three SNPs were genotyped based on previously determined functional and biological significance. We used multivariable Cox proportional hazards models to assess per-allele associations with the primary outcome defined as time to a composite endpoint including development of lethal prostate cancer or biochemical recurrence. RESULTS We followed 434 patients treated with radiation therapy for a median of 9 years. On multivariate analysis, the rs12757998 variant allele was associated with significantly decreased risk of the composite endpoint [HR: 0.65; 95% confidence interval (CI), 0.45-0.94%; P = 0.02] driven by decreased biochemical recurrence (HR: 0.60; 95% CI, 0.40-0.89%; P = 0.01) and men treated with external beam (HR: 0.58; 95% CI, 0.36-0.93%; P = 0.02). In contrast, in 516 men from the same cohort treated with radical prostatectomy, we found no significant impact of this variant on outcome. Furthermore, the rs12757998 variant allele significantly modified the association between androgen deprivation therapy and outcomes after radiation therapy (p-interaction = 0.02). CONCLUSION We show an association between RNASEL SNP rs12757998 and outcome after radiation therapy for prostate cancer. This SNP is associated with increased circulating C-reactive protein and interleukin-6, suggesting a potential role for inflammation in the response to radiation. If validated, genetic predictors of outcome may help inform prostate cancer management.
Collapse
|
12
|
Bailey-Wilson JE, Childs EJ, Cropp CD, Schaid DJ, Xu J, Camp NJ, Cannon-Albright LA, Farnham JM, George A, Powell I, Carpten JD, Giles GG, Hopper JL, Severi G, English DR, Foulkes WD, Mæhle L, Møller P, Eeles R, Easton D, Guy M, Edwards S, Badzioch MD, Whittemore AS, Oakley-Girvan I, Hsieh CL, Dimitrov L, Stanford JL, Karyadi DM, Deutsch K, McIntosh L, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Thibodeau SN, McDonnell SK, Hebbring S, Lange EM, Cooney KA, Tammela TLJ, Schleutker J, Maier C, Bochum S, Hoegel J, Grönberg H, Wiklund F, Emanuelsson M, Cancel-Tassin G, Valeri A, Cussenot O, Isaacs WB. Analysis of Xq27-28 linkage in the international consortium for prostate cancer genetics (ICPCG) families. BMC MEDICAL GENETICS 2012; 13:46. [PMID: 22712434 PMCID: PMC3495053 DOI: 10.1186/1471-2350-13-46] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 04/30/2012] [Indexed: 11/20/2022]
Abstract
BACKGROUND Genetic variants are likely to contribute to a portion of prostate cancer risk. Full elucidation of the genetic etiology of prostate cancer is difficult because of incomplete penetrance and genetic and phenotypic heterogeneity. Current evidence suggests that genetic linkage to prostate cancer has been found on several chromosomes including the X; however, identification of causative genes has been elusive. METHODS Parametric and non-parametric linkage analyses were performed using 26 microsatellite markers in each of 11 groups of multiple-case prostate cancer families from the International Consortium for Prostate Cancer Genetics (ICPCG). Meta-analyses of the resultant family-specific linkage statistics across the entire 1,323 families and in several predefined subsets were then performed. RESULTS Meta-analyses of linkage statistics resulted in a maximum parametric heterogeneity lod score (HLOD) of 1.28, and an allele-sharing lod score (LOD) of 2.0 in favor of linkage to Xq27-q28 at 138 cM. In subset analyses, families with average age at onset less than 65 years exhibited a maximum HLOD of 1.8 (at 138 cM) versus a maximum regional HLOD of only 0.32 in families with average age at onset of 65 years or older. Surprisingly, the subset of families with only 2-3 affected men and some evidence of male-to-male transmission of prostate cancer gave the strongest evidence of linkage to the region (HLOD = 3.24, 134 cM). For this subset, the HLOD was slightly increased (HLOD = 3.47 at 134 cM) when families used in the original published report of linkage to Xq27-28 were excluded. CONCLUSIONS Although there was not strong support for linkage to the Xq27-28 region in the complete set of families, the subset of families with earlier age at onset exhibited more evidence of linkage than families with later onset of disease. A subset of families with 2-3 affected individuals and with some evidence of male to male disease transmission showed stronger linkage signals. Our results suggest that the genetic basis for prostate cancer in our families is much more complex than a single susceptibility locus on the X chromosome, and that future explorations of the Xq27-28 region should focus on the subset of families identified here with the strongest evidence of linkage to this region.
Collapse
Affiliation(s)
- Joan E Bailey-Wilson
- Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, 21224, USA
- African American Hereditary Prostate Cancer ICPCG Group, Phoenix, AZ, USA
- University of Tampere ICPCG Group, Tampere, Finland
| | - Erica J Childs
- Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, 21224, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Cheryl D Cropp
- Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jianfeng Xu
- Data Coordinating Center for the ICPCG and Center for Human Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Nicola J Camp
- University of Utah ICPCG Group and Division of Genetic Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Lisa A Cannon-Albright
- University of Utah ICPCG Group and Division of Genetic Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - James M Farnham
- University of Utah ICPCG Group and Division of Genetic Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Asha George
- Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, 21224, USA
- African American Hereditary Prostate Cancer ICPCG Group, Phoenix, AZ, USA
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Isaac Powell
- African American Hereditary Prostate Cancer ICPCG Group, Phoenix, AZ, USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - John D Carpten
- African American Hereditary Prostate Cancer ICPCG Group, Phoenix, AZ, USA
- Translational Genomics Research Institute, Genetic Basis of Human Disease Research Division, Phoenix, AZ, USA
| | - Graham G Giles
- ACTANE consortium
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, The University of Melbourne, Melbourne, Australia
| | - John L Hopper
- ACTANE consortium
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Gianluca Severi
- ACTANE consortium
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Dallas R English
- ACTANE consortium
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, The University of Melbourne, Melbourne, Australia
| | - William D Foulkes
- ACTANE consortium
- Program in Cancer Genetics, McGill University, Montreal, QC, Canada
| | - Lovise Mæhle
- ACTANE consortium
- Department of Medical Genetics, Oslo University Hospital, The Norwegian Radium Hospital, Oslo,Norway
| | - Pål Møller
- ACTANE consortium
- Department of Medical Genetics, Oslo University Hospital, The Norwegian Radium Hospital, Oslo,Norway
| | - Rosalind Eeles
- ACTANE consortium
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Douglas Easton
- ACTANE consortium
- Cancer Research UK Genetic Epidemiology Unit, Cambridge, UK
| | - Michelle Guy
- ACTANE consortium
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Steve Edwards
- ACTANE consortium
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Michael D Badzioch
- ACTANE consortium
- Division of Medical Genetics, University of Washington Medical Center, Seattle, WA, USA
| | - Alice S Whittemore
- BC/CA/HI ICPCG Group, Stanford, CA, USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Ingrid Oakley-Girvan
- BC/CA/HI ICPCG Group, Stanford, CA, USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
- Cancer Prevention Institute of California
| | - Chih-Lin Hsieh
- BC/CA/HI ICPCG Group, Stanford, CA, USA
- Department of Urology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Ageles, CA, USA
| | - Latchezar Dimitrov
- Data Coordinating Center for the ICPCG and Center for Human Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Janet L Stanford
- FHCRC ICPCG Group, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, USA
| | - Danielle M Karyadi
- FHCRC ICPCG Group, Seattle, WA, USA
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kerry Deutsch
- FHCRC ICPCG Group, Seattle, WA, USA
- Institute for Systems Biology, Seattle, WA, USA
| | - Laura McIntosh
- FHCRC ICPCG Group, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, USA
| | - Elaine A Ostrander
- FHCRC ICPCG Group, Seattle, WA, USA
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen E Wiley
- Johns Hopkins University ICPCG Group and Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Sarah D Isaacs
- Johns Hopkins University ICPCG Group and Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Patrick C Walsh
- Johns Hopkins University ICPCG Group and Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | - Ethan M Lange
- University of Michigan ICPCG Group, Ann Arbor, MI, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Kathleen A Cooney
- University of Michigan ICPCG Group, Ann Arbor, MI, USA
- University of Michigan, Ann Arbor, MI, USA
| | - Teuvo LJ Tammela
- University of Tampere ICPCG Group, Tampere, Finland
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- Centre for Laboratory Medicine and Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Johanna Schleutker
- University of Tampere ICPCG Group, Tampere, Finland
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- Centre for Laboratory Medicine and Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Christiane Maier
- University of Ulm ICPCG Group, Ulm, Germany
- Dept of Urology, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Sylvia Bochum
- University of Ulm ICPCG Group, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Josef Hoegel
- University of Ulm ICPCG Group, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Olivier Cussenot
- CeRePP ICPCG Group, 75020, Paris, France
- Hopital Tenon, Assistance Publique-Hopitaux de Paris, 75020, Paris, France
| | - William B Isaacs
- Johns Hopkins University ICPCG Group and Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|