1
|
Bray AW, Duan R, Malalur P, Drusbosky LM, Gourdin TS, Hill EG, Lilly MB. Elevated serum CEA is associated with liver metastasis and distinctive circulating tumor DNA alterations in patients with castration-resistant prostate cancer. Prostate 2022; 82:1264-1272. [PMID: 35766303 PMCID: PMC9388585 DOI: 10.1002/pros.24400] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/14/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Elevated serum carcinoembryonic antigen (CEA) is used to identify "treatment emergent" forms of castration-resistant prostate cancer (CRPC) such as aggressive variant prostate cancer (AVPC). However, its individual utility as a prognostic marker and the genetic alterations associated with its expression have not been extensively studied in CRPC. METHODS This study retrospectively analyzed clinical outcomes and circulating tumor DNA profiles in 163 patients with CRPC and elevated or normal serum CEA. These same patients were then classified as AVPC or non-AVPC and compared to determine the uniqueness of CEA-associated gene alterations. RESULTS Patients with elevated CEA demonstrated higher rates of liver metastasis (37.5% vs. 19.1%, p = 0.02) and decreased median overall survival from CRPC diagnosis (28.7 vs. 73.2 mo, p < 0.0001). In addition, patients with elevated CEA were more likely to harbor copy number amplifications (CNAs) in AR, PIK3CA, MYC, BRAF, CDK6, MET, CCNE1, KIT, RAF1, and KRAS. Based on variant allele frequency we also defined "clonal" single-nucleotide variants (SNVs) thought to be driving disease progression in each patient and found that CEA expression was negatively correlated with clonal AR SNVs and positively correlated with clonal TP53 SNVs. Of these genetic associations, only the increases in clonal TP53 SNVs and KRAS amplifications were recapitulated among patients with AVPC when compared to patients without AVPC. CONCLUSIONS Together these findings suggest that CEA expression in CRPC is associated with aggressive clinical behavior and gene alterations distinct from those in AVPC.
Collapse
Affiliation(s)
- Alexander W. Bray
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Rong Duan
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Pannaga Malalur
- The Ohio State University Wexner Medical CenterColumbusOhioUSA
| | | | - Theodore S. Gourdin
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Elizabeth G. Hill
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Michael B. Lilly
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
2
|
Graf RP, Eskander R, Brueggeman L, Stupack DG. Association of Copy Number Variation Signature and Survival in Patients With Serous Ovarian Cancer. JAMA Netw Open 2021; 4:e2114162. [PMID: 34181012 PMCID: PMC8239953 DOI: 10.1001/jamanetworkopen.2021.14162] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IMPORTANCE Tailoring therapeutic regimens to individual patients with ovarian cancer is informed by severity of disease using a variety of clinicopathologic indicators. Although DNA repair variations are increasingly used for therapy selection in ovarian cancer, molecular features are not widely used for general assessment of patient prognosis and disease severity. OBJECTIVE To distill a highly dynamic characteristic, signature of copy number variations (CNV), into a risk score that could be easily validated analytically or repurposed for use given existing US Food and Drug Administration (FDA)-approved multigene assays. DESIGN, SETTING, AND PARTICIPANTS This genetic association study used the Cancer Genome Atlas Ovarian Cancer database to assess for genome-wide survival associations agnostic to gene function. Regions enriched for significant associations were compared to associations from scrambled data. CNV associations were condensed into a risk score, which was internally validated using bootstrapping. The participants were patients with serous ovarian cancer (stages I-IV) diagnosed from 1992 to 2013. Statistical analysis was performed from April to July 2020. MAIN OUTCOMES AND MEASURES Overall survival (OS). RESULTS Among 564 patients with serous ovarian cancer, the mean (SD) age was 59.7 (11.5) years; 34 (6%) identified as Black or African American. A total of 13 genome regions, comprising 14 alterations, were identified as significantly risk associated. Composite risk score was independent of total CNV burden, total mutational burden, BRCA status, and open-source genome-wide DNA repair deficiency signatures. Binned terciles yielded high-, standard-, and low-risk groups with respective median OS estimates of 2.9 (95% CI, 2.3-3.2) years, 4.1 (95% CI, 3.7-4.8) years, and 5.7 (95% CI, 4.7-7.4) years, respectively (P < .001). Associated 5-year survival estimates in each tercile were 15% (95% CI, 10%-22%), 36% (95% CI, 29%-46%), and 53% (95% CI, 45%-62%). The risk score had more discriminatory ability to prognosticate OS than age, clinical stage, grade, and race combined, and was strongly additive to significant clinical features (P < .001). Simulated adaptation of FDA-approved assays showed similar performance. Gene ontology analyses of identified regions showed an enrichment for regulatory miRNAs and protein kinase regulators. CONCLUSIONS AND RELEVANCE This study found that a CNV-based risk score is independent to and stronger than current or near-future ovarian cancer genomic biomarkers to prognosticate OS. CNV regions identified were not strongly associated with canonical ovarian cancer biological pathways, identifying candidates for future mechanistic investigations. External validation of the CNV risk score, especially in concert with more extensive clinical features, could be pursued via existing FDA-approved assays.
Collapse
Affiliation(s)
- Ryon P. Graf
- Moores Cancer Center, University of California, San Diego
- Now at Foundation Medicine Inc, Cambridge, Massachusetts
| | - Ramez Eskander
- Moores Cancer Center, University of California, San Diego
| | - Leo Brueggeman
- Interdisciplinary Genetics Program, University of Iowa, Iowa City
- Medical Scientist Training Program, University of Iowa, Iowa City
| | | |
Collapse
|
3
|
Saunders EJ, Kote-Jarai Z, Eeles RA. Identification of Germline Genetic Variants that Increase Prostate Cancer Risk and Influence Development of Aggressive Disease. Cancers (Basel) 2021; 13:760. [PMID: 33673083 PMCID: PMC7917798 DOI: 10.3390/cancers13040760] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PrCa) is a heterogeneous disease, which presents in individual patients across a diverse phenotypic spectrum ranging from indolent to fatal forms. No robust biomarkers are currently available to enable routine screening for PrCa or to distinguish clinically significant forms, therefore late stage identification of advanced disease and overdiagnosis plus overtreatment of insignificant disease both remain areas of concern in healthcare provision. PrCa has a substantial heritable component, and technological advances since the completion of the Human Genome Project have facilitated improved identification of inherited genetic factors influencing susceptibility to development of the disease within families and populations. These genetic markers hold promise to enable improved understanding of the biological mechanisms underpinning PrCa development, facilitate genetically informed PrCa screening programmes and guide appropriate treatment provision. However, insight remains largely lacking regarding many aspects of their manifestation; especially in relation to genes associated with aggressive phenotypes, risk factors in non-European populations and appropriate approaches to enable accurate stratification of higher and lower risk individuals. This review discusses the methodology used in the elucidation of genetic loci, genes and individual causal variants responsible for modulating PrCa susceptibility; the current state of understanding of the allelic spectrum contributing to PrCa risk; and prospective future translational applications of these discoveries in the developing eras of genomics and personalised medicine.
Collapse
Affiliation(s)
- Edward J. Saunders
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
| | - Rosalind A. Eeles
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
- Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| |
Collapse
|
4
|
Innis SM, Cabot B. GBAF, a small BAF sub-complex with big implications: a systematic review. Epigenetics Chromatin 2020; 13:48. [PMID: 33143733 PMCID: PMC7607862 DOI: 10.1186/s13072-020-00370-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022] Open
Abstract
ATP-dependent chromatin remodeling by histone-modifying enzymes and chromatin remodeling complexes is crucial for maintaining chromatin organization and facilitating gene transcription. In the SWI/SNF family of ATP-dependent chromatin remodelers, distinct complexes such as BAF, PBAF, GBAF, esBAF and npBAF/nBAF are of particular interest regarding their implications in cellular differentiation and development, as well as in various diseases. The recently identified BAF subcomplex GBAF is no exception to this, and information is emerging linking this complex and its components to crucial events in mammalian development. Furthermore, given the essential nature of many of its subunits in maintaining effective chromatin remodeling function, it comes as no surprise that aberrant expression of GBAF complex components is associated with disease development, including neurodevelopmental disorders and numerous malignancies. It becomes clear that building upon our knowledge of GBAF and BAF complex function will be essential for advancements in both mammalian reproductive applications and the development of more effective therapeutic interventions and strategies. Here, we review the roles of the SWI/SNF chromatin remodeling subcomplex GBAF and its subunits in mammalian development and disease.
Collapse
Affiliation(s)
- Sarah M Innis
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Birgit Cabot
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
5
|
Darst BF, Dadaev T, Saunders E, Sheng X, Wan P, Pooler L, Xia LY, Chanock S, Berndt SI, Gapstur SM, Stevens V, Albanes D, Weinstein SJ, Gnanapragasam V, Giles GG, Nguyen-Dumont T, Milne RL, Pomerantz M, Schmidt JA, Mucci L, Catalona WJ, Hetrick KN, Doheny KF, MacInnis RJ, Southey MC, Eeles RA, Wiklund F, Kote-Jarai Z, Conti DV, Haiman CA. Germline Sequencing DNA Repair Genes in 5545 Men With Aggressive and Nonaggressive Prostate Cancer. J Natl Cancer Inst 2020; 113:616-625. [PMID: 32853339 DOI: 10.1093/jnci/djaa132] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/27/2020] [Accepted: 08/20/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND There is an urgent need to identify factors specifically associated with aggressive prostate cancer (PCa) risk. We investigated whether rare pathogenic, likely pathogenic, or deleterious (P/LP/D) germline variants in DNA repair genes are associated with aggressive PCa risk in a case-case study of aggressive vs nonaggressive disease. METHODS Participants were 5545 European-ancestry men, including 2775 nonaggressive and 2770 aggressive PCa cases, which included 467 metastatic cases (16.9%). Samples were assembled from 12 international studies and germline sequenced together. Rare (minor allele frequency < 0.01) P/LP/D variants were analyzed for 155 DNA repair genes. We compared single variant, gene-based, and DNA repair pathway-based burdens by disease aggressiveness. All statistical tests are 2-sided. RESULTS BRCA2 and PALB2 had the most statistically significant gene-based associations, with 2.5% of aggressive and 0.8% of nonaggressive cases carrying P/LP/D BRCA2 alleles (odds ratio [OR] = 3.19, 95% confidence interval [CI] = 1.94 to 5.25, P = 8.58 × 10-7) and 0.65% of aggressive and 0.11% of nonaggressive cases carrying P/LP/D PALB2 alleles (OR = 6.31, 95% CI = 1.83 to 21.68, P = 4.79 × 10-4). ATM had a nominal association, with 1.6% of aggressive and 0.8% of nonaggressive cases carrying P/LP/D ATM alleles (OR = 1.88, 95% CI = 1.10 to 3.22, P = .02). In aggregate, P/LP/D alleles within 24 literature-curated candidate PCa DNA repair genes were more common in aggressive than nonaggressive cases (carrier frequencies = 14.2% vs 10.6%, respectively; P = 5.56 × 10-5). However, this difference was non-statistically significant (P = .18) on excluding BRCA2, PALB2, and ATM. Among these 24 genes, P/LP/D carriers had a 1.06-year younger diagnosis age (95% CI = -1.65 to 0.48, P = 3.71 × 10-4). CONCLUSIONS Risk conveyed by DNA repair genes is largely driven by rare P/LP/D alleles within BRCA2, PALB2, and ATM. These findings support the importance of these genes in both screening and disease management considerations.
Collapse
Affiliation(s)
- Burcu F Darst
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Ed Saunders
- The Institute of Cancer Research, London, UK
| | - Xin Sheng
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peggy Wan
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Loreall Pooler
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lucy Y Xia
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Chanock
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sonja I Berndt
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Demetrius Albanes
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Vincent Gnanapragasam
- Department of Surgery and Oncology, Academic Urology Group, University of Cambridge, Cambridge, UK
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia
| | - Tu Nguyen-Dumont
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Victoria, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia
| | | | | | | | | | - Kurt N Hetrick
- Department of Genetic Medicine, Center for Inherited Disease Research, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kimberly F Doheny
- Department of Genetic Medicine, Center for Inherited Disease Research, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert J MacInnis
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Victoria, Australia
| | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK.,The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | - David V Conti
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Zhou Q, Xie Y, Wang L, Xu T, Gao Y. LncRNA EWSAT1 upregulates CPEB4 via miR-330-5p to promote cervical cancer development. Mol Cell Biochem 2020; 471:177-188. [PMID: 32556917 DOI: 10.1007/s11010-020-03778-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022]
Abstract
Long non-coding RNA (lncRNA) Ewing sarcoma associated transcript 1 (EWSAT1) is an oncogene in a variety of tumors. Here, we planned to demonstrate EWSAT1 function in cervical cancer and further illustrate its underlying mechanism. EWSAT1 expression in cervical cancer was evaluated through qRT-PCR. Colony forming capacity was measured by colony formation assay and cell proliferation ability was measured by CCK-8 kit. Wound healing experiment was applied to test the cell migration and transwell assay was applied to test the invasion ability. Luciferase assay was employed to demonstrate EWSAT1 and miR-330-5p interaction. In cervical cancer, the expression of EWSAT1 was enhanced and contributed to the poor prognosis. Downregulated EWSAT1 expression inhibited Hela cell migration, proliferation, and invasion. EWSAT1 targeted to miR-330-5p and upregulated cytoplasmic polyadenylation element-binding protein 4 (CPEB4) expression by sponging miR-330-5p. Our study revealed that EWSAT1 enhances CPEB4 expression through sponging miR-330-5p, thereby promoting cervical cancer development, which might provide potential therapeutic targets for clinically cervical cancer patients.
Collapse
Affiliation(s)
- Qingyan Zhou
- Department of Gynecology, Dongchangfu District, Liaocheng Dongchangfu People's Hospital, No. 128 Songgui Road, Liaocheng, 252000, Shandong, China.
| | - Yuan Xie
- Department of Gynecology, Dongchangfu District, Liaocheng Dongchangfu People's Hospital, No. 128 Songgui Road, Liaocheng, 252000, Shandong, China
| | - Li Wang
- Department of Gynecology, Dongchangfu District, Liaocheng Dongchangfu People's Hospital, No. 128 Songgui Road, Liaocheng, 252000, Shandong, China
| | - Tao Xu
- Delivery Room, Dongchangfu District, Liaocheng Dongchangfu People's Hospital, No. 128 Songgui Road, Liaocheng, 252000, Shandong, China
| | - Yongbin Gao
- Department of Gynecology, Dongchangfu District, Liaocheng Dongchangfu People's Hospital, No. 128 Songgui Road, Liaocheng, 252000, Shandong, China
| |
Collapse
|
7
|
Circulating microRNAs as potential diagnostic biomarkers and therapeutic targets in prostate cancer: Current status and future perspectives. J Cell Biochem 2019; 120:16316-16329. [DOI: 10.1002/jcb.29053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/04/2019] [Indexed: 12/19/2022]
|
8
|
Ma X, Du T, Zhu D, Chen X, Lai Y, Wu W, Wang Q, Lin C, Li Z, Liu L, Huang H. High levels of glioma tumor suppressor candidate region gene 1 predicts a poor prognosis for prostate cancer. Oncol Lett 2018; 16:6749-6755. [PMID: 30405818 DOI: 10.3892/ol.2018.9490] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/08/2018] [Indexed: 11/06/2022] Open
Abstract
Glioma tumor suppressor candidate region gene 1 (GLTSCR1) is associated with the progression of oligodendroglioma. However, there has been little study of GLTSCR1 in prostate cancer. In the present study, the association between the expression of GLTSCR1, and the progression and prognosis of tumors in patients with prostate cancer was assessed. An immunohistochemical analysis was performed using a human tissue microarray for GLTSCR1 at the protein expression level and the immunostaining results were evaluated against clinical variables of patients with prostate cancer. Subsequently, The Cancer Genome Atlas (TCGA) was used to validate the analysis results at the mRNA level and to study the prognostic value of GLTSCR1 in prostate cancer. Immunohistochemistry and TCGA data analysis revealed that GLTSCR1 expression in the prostate cancer tissues was significantly higher than that in the benign prostate tissues (immunoreactivity score, P=0.015; mRNA levels: cancer, 447.7±6.45 vs. benign, 343.5±4.21; P<0.001). Additionally, the increased GLTSCR1 protein expression was associated with certain clinical variables in the prostate cancer tissues, including advanced clinical stage (P<0.001), enhanced tumor invasion (P=0.003), lymph node metastasis (P=0.003) and distant metastasis (P=0.001). TCGA data revealed similar results, demonstrating that the upregulation of GLTSCR1 mRNA expression was associated with the Gleason score (P<0.001), enhanced tumor invasion (P=0.011), lymph node metastasis (P=0.001) and distant metastasis (P=0.002). Furthermore, Kaplan-Meier analysis suggested that among all patients, high GLTSCR1 expression indicated a decreased overall survival (P=0.028) and biochemical recurrence (BCR)-free survival (P=0.004), compared with patients with low GLTSCR1 expression. Finally, multivariate analysis revealed that the expression of GLTSCR1 was an independent predictor of poor BCR-free survival (P=0.049). The present study suggested that the increased expression of GLTSCR1 was associated with the progression of prostate cancer. Furthermore, GLTSCR1 may be a novel biomarker that is able to predict the clinical outcome in prostate cancer patients.
Collapse
Affiliation(s)
- Xiaoming Ma
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Tao Du
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Dingjun Zhu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xianju Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yiming Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wanhua Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Qiong Wang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chunhao Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zean Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Leyuan Liu
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA.,Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Hai Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
9
|
Tinay I, Tan M, Gui B, Werner L, Kibel AS, Jia L. Functional roles and potential clinical application of miRNA-345-5p in prostate cancer. Prostate 2018; 78:927-937. [PMID: 29748958 DOI: 10.1002/pros.23650] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNAs, which negatively regulate gene expression and impact prostate cancer (PCa) growth and progression. Circulating miRNAs are stable and detectable in cell-free body fluids, such as serum. Investigation of circulating miRNAs presents great potential in uncovering new insights into the roles of miRNAs in PCa diagnosis and therapy. METHODS Using TaqMan miRNA quantitative reverse transcription polymerase chain reaction (RT-qPCR), we compared the expression levels of five miRNAs (miR-193a-3p, miR-9-3p, miR-335-5p, miR-330-3p, and miR-345-5p) in serum samples from 20 normal individuals without cancer, 25 patients with localized disease, 25 patients with hormone-naïve or hormone sensitive metastatic disease, and 25 patients with metastatic castration-resistant prostate cancer (CRPC). These five miRNAs were identified as potential oncogenes in our previous studies. MiR-345-5p was further investigated for its functional roles in CRPC cells. RESULTS We discovered that miR-9-3p, miR-330-3p-3p, and miR-345-5p were significantly overexpressed in serum from PCa patients when compared to serum from individuals without cancer. No differential expression patterns were observed between different disease categories. However, patients who were in remission after androgen deprivation therapy (ADT) appeared to have significantly lower miR-345-5p levels compared to the rest of the groups. We further demonstrated that miR-345-5p promotes CRPC cell growth and migration in vitro and validated that CDKN1A (the gene encoding p21) is the direct target of miR-345-5p. CONCLUSIONS Our results set the stage for a further investigation on the potential application of circulating miR-345-5p as a biomarker for PCa diagnosis and therapeutic response. The oncogenic roles of miR-345-5p through targeting CDKN1A render it a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Ilker Tinay
- Division of Urology, Department of Surgery, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Urology, Marmara University School of Medicine, Istanbul, Turkey
| | - Mingyue Tan
- Division of Urology, Department of Surgery, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Bin Gui
- Division of Urology, Department of Surgery, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lillian Werner
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adam S Kibel
- Division of Urology, Department of Surgery, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Li Jia
- Division of Urology, Department of Surgery, Brigham Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Abstract
Although prostate cancer is the most common malignancy to affect men in the Western world, the molecular mechanisms underlying its development and progression remain poorly understood. Like all cancers, prostate cancer is a genetic disease that is characterized by multiple genomic alterations, including point mutations, microsatellite variations, and chromosomal alterations such as translocations, insertions, duplications, and deletions. In prostate cancer, but not other carcinomas, these chromosome alterations result in a high frequency of gene fusion events. The development and application of novel high-resolution technologies has significantly accelerated the detection of genomic alterations, revealing the complex nature and heterogeneity of the disease. The clinical heterogeneity of prostate cancer can be partly explained by this underlying genetic heterogeneity, which has been observed between patients from different geographical and ethnic populations, different individuals within these populations, different tumour foci within the same patient, and different cells within the same tumour focus. The highly heterogeneous nature of prostate cancer provides a real challenge for clinical disease management and a detailed understanding of the genetic alterations in all cells, including small subpopulations, would be highly advantageous.
Collapse
|
11
|
Abstract
Adhesion G protein-coupled receptors (aGPCRs) have a long evolutionary history dating back to very basal unicellular eukaryotes. Almost every vertebrate is equipped with a set of different aGPCRs. Genomic sequence data of several hundred extinct and extant species allows for reconstruction of aGPCR phylogeny in vertebrates and non-vertebrates in general but also provides a detailed view into the recent evolutionary history of human aGPCRs. Mining these sequence sources with bioinformatic tools can unveil many facets of formerly unappreciated aGPCR functions. In this review, we extracted such information from the literature and open public sources and provide insights into the history of aGPCR in humans. This includes comprehensive analyses of signatures of selection, variability of human aGPCR genes, and quantitative traits at human aGPCR loci. As indicated by a large number of genome-wide genotype-phenotype association studies, variations in aGPCR contribute to specific human phenotypes. Our survey demonstrates that aGPCRs are significantly involved in adaptation processes, phenotype variations, and diseases in humans.
Collapse
Affiliation(s)
- Peter Kovacs
- Integrated Research and Treatment Center (IFB) AdiposityDiseases, Medical Faculty, University of Leipzig, Liebigstr. 21, Leipzig, 04103, Germany.
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
| |
Collapse
|
12
|
Helfand BT, Roehl KA, Cooper PR, McGuire BB, Fitzgerald LM, Cancel-Tassin G, Cornu JN, Bauer S, Van Blarigan EL, Chen X, Duggan D, Ostrander EA, Gwo-Shu M, Zhang ZF, Chang SC, Jeong S, Fontham ETH, Smith G, Mohler JL, Berndt SI, McDonnell SK, Kittles R, Rybicki BA, Freedman M, Kantoff PW, Pomerantz M, Breyer JP, Smith JR, Rebbeck TR, Mercola D, Isaacs WB, Wiklund F, Cussenot O, Thibodeau SN, Schaid DJ, Cannon-Albright L, Cooney KA, Chanock SJ, Stanford JL, Chan JM, Witte J, Xu J, Bensen JT, Taylor JA, Catalona WJ. Associations of prostate cancer risk variants with disease aggressiveness: results of the NCI-SPORE Genetics Working Group analysis of 18,343 cases. Hum Genet 2015; 134:439-50. [PMID: 25715684 PMCID: PMC4586077 DOI: 10.1007/s00439-015-1534-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/06/2015] [Indexed: 01/18/2023]
Abstract
Genetic studies have identified single nucleotide polymorphisms (SNPs) associated with the risk of prostate cancer (PC). It remains unclear whether such genetic variants are associated with disease aggressiveness. The NCI-SPORE Genetics Working Group retrospectively collected clinicopathologic information and genotype data for 36 SNPs which at the time had been validated to be associated with PC risk from 25,674 cases with PC. Cases were grouped according to race, Gleason score (Gleason ≤ 6, 7, ≥ 8) and aggressiveness (non-aggressive, intermediate, and aggressive disease). Statistical analyses were used to compare the frequency of the SNPs between different disease cohorts. After adjusting for multiple testing, only PC-risk SNP rs2735839 (G) was significantly and inversely associated with aggressive (OR = 0.77; 95 % CI 0.69-0.87) and high-grade disease (OR = 0.77; 95 % CI 0.68-0.86) in European men. Similar associations with aggressive (OR = 0.72; 95 % CI 0.58-0.89) and high-grade disease (OR = 0.69; 95 % CI 0.54-0.87) were documented in African-American subjects. The G allele of rs2735839 was associated with disease aggressiveness even at low PSA levels (<4.0 ng/mL) in both European and African-American men. Our results provide further support that a PC-risk SNP rs2735839 near the KLK3 gene on chromosome 19q13 may be associated with aggressive and high-grade PC. Future prospectively designed, case-case GWAS are needed to identify additional SNPs associated with PC aggressiveness.
Collapse
Affiliation(s)
- Brian T Helfand
- Department of Surgery, Division of Urology, John and Carol Walter Center for Urological Health, NorthShore University Health System, Evanston, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Decker B, Ostrander EA. Dysregulation of the homeobox transcription factor gene HOXB13: role in prostate cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:193-201. [PMID: 25206306 PMCID: PMC4157396 DOI: 10.2147/pgpm.s38117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Prostate cancer (PC) is the most common noncutaneous cancer in men, and epidemiological studies suggest that about 40% of PC risk is heritable. Linkage analyses in hereditary PC families have identified multiple putative loci. However, until recently, identification of specific risk alleles has proven elusive. Cooney et al used linkage mapping and segregation analysis to identify a putative risk locus on chromosome 17q21-22. In search of causative variant(s) in genes from the candidate region, a novel, potentially deleterious G84E substitution in homeobox transcription factor gene HOXB13 was observed in multiple hereditary PC families. In follow-up testing, the G84E allele was enriched in cases, especially those with an early diagnosis or positive family history of disease. This finding was replicated by others, confirming HOXB13 as a PC risk gene. The HOXB13 protein plays diverse biological roles in embryonic development and terminally differentiated tissue. In tumor cell lines, HOXB13 participates in a number of biological functions, including coactivation and localization of the androgen receptor and FOXA1. However, no consensus role has emerged and many questions remain. All HOXB13 variants with a proposed role in PC risk are predicted to damage the protein and lie in domains that are highly conserved across species. The G84E variant has the strongest epidemiological support and lies in a highly conserved MEIS protein-binding domain, which binds cofactors required for activation. On the basis of epidemiological and biological data, the G84E variant likely modulates the interaction between the HOXB13 protein and the androgen receptor, as well as affecting FOXA1-mediated transcriptional programming. However, further studies of the mutated protein are required to clarify the mechanisms by which this translates into PC risk.
Collapse
Affiliation(s)
- Brennan Decker
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA ; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Mao Y, Chen H, Lin Y, Xu X, Hu Z, Zhu Y, Wu J, Xu X, Zheng X, Xie L. microRNA-330 inhibits cell motility by downregulating Sp1 in prostate cancer cells. Oncol Rep 2013; 30:327-33. [PMID: 23670210 DOI: 10.3892/or.2013.2452] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/16/2013] [Indexed: 12/26/2022] Open
Abstract
microRNAs (miRNAs), small non-coding RNAs, have emerged as key regulators of a large number of genes. The present study aimed to explore novel biological functions of miR-330 in the human prostate cancer cell lines DU145 and PC3. We confirmed that miR-330 was downregulated and inversely correlated with specificity protein 1 (Sp1) expression. Overexpression of miR-330 by transfection of a chemically synthesized miR-330 mimic induced a reduction in expression levels of the Sp1 protein, accompanied by significant suppression of cellular migration and invasion capability. In addition, the Sp1-knockdown experiments presented similar phenomena. Finally, the luciferase reporter assay validated Sp1 as the direct target of miR-330. These findings indicate that miR-330 acts as an anti-metastatic miRNA in prostate cancer.
Collapse
Affiliation(s)
- Yeqing Mao
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Amin Al Olama A, Kote-Jarai Z, Schumacher FR, Wiklund F, Berndt SI, Benlloch S, Giles GG, Severi G, Neal DE, Hamdy FC, Donovan JL, Hunter DJ, Henderson BE, Thun MJ, Gaziano M, Giovannucci EL, Siddiq A, Travis RC, Cox DG, Canzian F, Riboli E, Key TJ, Andriole G, Albanes D, Hayes RB, Schleutker J, Auvinen A, Tammela TL, Weischer M, Stanford JL, Ostrander EA, Cybulski C, Lubinski J, Thibodeau SN, Schaid DJ, Sorensen KD, Batra J, Clements JA, Chambers S, Aitken J, Gardiner RA, Maier C, Vogel W, Dörk T, Brenner H, Habuchi T, Ingles S, John EM, Dickinson JL, Cannon-Albright L, Teixeira MR, Kaneva R, Zhang HW, Lu YJ, Park JY, Cooney KA, Muir KR, Leongamornlert DA, Saunders E, Tymrakiewicz M, Mahmud N, Guy M, Govindasami K, O'Brien LT, Wilkinson RA, Hall AL, Sawyer EJ, Dadaev T, Morrison J, Dearnaley DP, Horwich A, Huddart RA, Khoo VS, Parker CC, Van As N, Woodhouse CJ, Thompson A, Dudderidge T, Ogden C, Cooper CS, Lophatonanon A, Southey MC, Hopper JL, English D, Virtamo J, Le Marchand L, Campa D, Kaaks R, Lindstrom S, Diver WR, Gapstur S, Yeager M, Cox A, Stern MC, Corral R, Aly M, Isaacs W, Adolfsson J, Xu J, Zheng SL, Wahlfors T, Taari K, Kujala P, Klarskov P, Nordestgaard BG, Røder MA, Frikke-Schmidt R, Bojesen SE, FitzGerald LM, Kolb S, Kwon EM, Karyadi DM, Orntoft TF, Borre M, Rinckleb A, Luedeke M, Herkommer K, Meyer A, Serth J, Marthick JR, Patterson B, Wokolorczyk D, Spurdle A, Lose F, McDonnell SK, Joshi AD, Shahabi A, Pinto P, Santos J, Ray A, Sellers TA, Lin HY, Stephenson RA, Teerlink C, Muller H, Rothenbacher D, Tsuchiya N, Narita S, Cao GW, Slavov C, Mitev V, Chanock S, Gronberg H, Haiman CA, Kraft P, Easton DF, Eeles RA. A meta-analysis of genome-wide association studies to identify prostate cancer susceptibility loci associated with aggressive and non-aggressive disease. Hum Mol Genet 2013; 22:408-15. [PMID: 23065704 PMCID: PMC3526158 DOI: 10.1093/hmg/dds425] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 10/04/2012] [Indexed: 01/14/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified multiple common genetic variants associated with an increased risk of prostate cancer (PrCa), but these explain less than one-third of the heritability. To identify further susceptibility alleles, we conducted a meta-analysis of four GWAS including 5953 cases of aggressive PrCa and 11 463 controls (men without PrCa). We computed association tests for approximately 2.6 million SNPs and followed up the most significant SNPs by genotyping 49 121 samples in 29 studies through the international PRACTICAL and BPC3 consortia. We not only confirmed the association of a PrCa susceptibility locus, rs11672691 on chromosome 19, but also showed an association with aggressive PrCa [odds ratio = 1.12 (95% confidence interval 1.03-1.21), P = 1.4 × 10(-8)]. This report describes a genetic variant which is associated with aggressive PrCa, which is a type of PrCa associated with a poorer prognosis.
Collapse
Affiliation(s)
- Ali Amin Al Olama
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Fredrick R. Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Sara Benlloch
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC 3053, Australia
| | - Gianluca Severi
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC 3053, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC 3053, Australia
| | - David E. Neal
- Surgical Oncology (Uro-Oncology: S4), Addenbrooke's Hospital, University of Cambridge, Box 279, Hills Road, Cambridge, UK
- Li Ka Shing Centre, Cancer Research UK Cambridge Research Institute, Cambridge CB2 2QQ, UK
| | - Freddie C. Hamdy
- Nuffield Department of Surgery and
- Faculty of Medical Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Jenny L. Donovan
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol BS8 2PS, UK
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Michael J. Thun
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Michael Gaziano
- Massachusetts Veterans Epidemiology and Research Information Center (MAVERIC) and Geriatric Research, Education, and Clinical Center (GRECC), Boston Veterans Affairs Healthcare System, Boston, MA 02114, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - David G. Cox
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Lyon Cancer Research Center, INSERM U1052, Lyon, France
| | | | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Timothy J. Key
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Gerald Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Richard B. Hayes
- Division of Epidemiology, Department of Environmental Medicine, NYU Langone Medical Centre, NYU Cancer Institute, New York, NY 10016, USA
| | - Johanna Schleutker
- Institute of Biomedical Technology/BioMediTech, University of Tampere and
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Anssi Auvinen
- Department of Epidemiology, School of Health Sciences and
| | - Teuvo L.J. Tammela
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | | | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Suzanne Chambers
- Griffith Health Institute, Griffith University, Gold Coast, QLD, Australia
- Viertel Centre for Research in Cancer Control, Cancer Council Queensland, Brisbane, QLD, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Joanne Aitken
- Viertel Centre for Research in Cancer Control, Cancer Council Queensland, Brisbane, QLD, Australia
| | - Robert A. Gardiner
- Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Christiane Maier
- Department of Urology and
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Walther Vogel
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Thilo Dörk
- Hannover Medical School, Hannover, Germany
| | | | - Tomonori Habuchi
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Sue Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, CA, USA
- Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Joanne L. Dickinson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Centre, Salt Lake City, UT, USA
| | - Manuel R. Teixeira
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Radka Kaneva
- Molecular Medicine Centre, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, 2 Zdrave St, Sofia 1431, Bulgaria
| | - Hong-Wei Zhang
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Yong-Jie Lu
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Jong Y. Park
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | - Kathleen A. Cooney
- Department of Internal Medicine and
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | - Edward Saunders
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Nadiya Mahmud
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Michelle Guy
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Koveela Govindasami
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Lynne T. O'Brien
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Amanda L. Hall
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Emma J. Sawyer
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Tokhir Dadaev
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Jonathan Morrison
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - David P. Dearnaley
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Alan Horwich
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Robert A. Huddart
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Vincent S. Khoo
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Christopher C. Parker
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Nicholas Van As
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | | | - Alan Thompson
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Tim Dudderidge
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Chris Ogden
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| | - Colin S. Cooper
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | | | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Grattan street, Parkville, VIC, Australia
| | - John L. Hopper
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC, Australia
| | - Dallas English
- Cancer Epidemiology Centre, Cancer Council Victoria, 1 Rathdowne Street, Carlton, VIC, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, 723 Swanston Street, Carlton, VIC, Australia
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Centre, Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Daniele Campa
- Lyon Cancer Research Center, INSERM U1052, Lyon, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Sara Lindstrom
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - W. Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Susan Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Angela Cox
- Department of Oncology, University of Sheffield, Sheffield, UK
| | - Mariana C. Stern
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Roman Corral
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
- Division of Urology, Department of Clinical Sciences, Danderyd Hospital and
| | - William Isaacs
- School of Medicine, Johns Hopkins University, 115 Marburg Building, 600 North Wolfe Street, Baltimore, MD 21205, USA
| | - Jan Adolfsson
- Oncological Centre, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Jianfeng Xu
- Center for Cancer Genomics, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - S. Lilly Zheng
- Center for Cancer Genomics, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Tiina Wahlfors
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | - Kimmo Taari
- Department of Urology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Paula Kujala
- Department of Pathology, Centre for Laboratory Medicine, Tampere University Hospital, Tampere, Finland
| | - Peter Klarskov
- Department of Urology, Herlev Hospital, Copenhagen University Hospital, Herlev Ringvej 75, Herlev DK-2730, Denmark
| | | | | | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen DK-2100, Denmark
| | | | - Liesel M. FitzGerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
| | - Erika M. Kwon
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | - Danielle M. Karyadi
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Room 5351, Bethesda, MD, USA
| | | | - Michael Borre
- Department of Urology, Aarhus University Hospital, Skejby, Denmark
| | | | - Manuel Luedeke
- Institute of Human Genetics, University Hospital Ulm, Ulm, Germany
| | - Kathleen Herkommer
- Department of Urology, Rechts der Isar Medical Centre, Technical University of Munich, Munich, Germany
| | | | | | - James R. Marthick
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Briony Patterson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS, Australia
| | - Dominika Wokolorczyk
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | | | - Felicity Lose
- Molecular Cancer Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | | | - Amit D. Joshi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Ahva Shahabi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Pedro Pinto
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Joana Santos
- Department of Genetics, Portuguese Oncology Institute and Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Ana Ray
- Department of Internal Medicine and
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Thomas A. Sellers
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | - Hui-Yi Lin
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, 12902 Magnolia Drive, Tampa, FL, USA
| | | | - Craig Teerlink
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Heiko Muller
- Division of Clinical Epidemiology and Aging Research and
| | | | - Norihiko Tsuchiya
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Shintaro Narita
- Department of Urology,Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| | - Guang-Wen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Chavdar Slavov
- Department of Urology and Alexandrovska University Hospital,Medical University of Sofia, Sofia, Bulgaria
| | - Vanio Mitev
- Molecular Medicine Centre, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, 2 Zdrave St, Sofia 1431, Bulgaria
| | | | | | | | | | - Stephen Chanock
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute, NIH, Gaithersburg, MD, USA
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 77, Sweden
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, CA, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology and
| | - Douglas F. Easton
- Strangeways Laboratory, Centre for Cancer Genetic Epidemiology, Worts Causeway, Cambridge CB1 8RN, UK
| | - Rosalind A. Eeles
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham and Sutton, London and Surrey, UK
| |
Collapse
|
16
|
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
|
17
|
Ledet EM, Sartor O, Rayford W, Bailey-Wilson JE, Mandal DM. Suggestive evidence of linkage identified at chromosomes 12q24 and 2p16 in African American prostate cancer families from Louisiana. Prostate 2012; 72:938-47. [PMID: 22615067 DOI: 10.1002/pros.21496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/13/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND In the United States, incidence of prostate cancer in African American men is more than twice than that of any other race. Thus far, numerous disease susceptibility loci have been identified for this cancer but definite locus-specific information is not yet established due to the tremendous amount of genetic and disease heterogeneity; additionally, despite high prevalence of prostate cancer amongst African American men, this population has been under represented in genetic studies of prostate cancer. METHODS In order to identify the susceptible locus (loci) for prostate cancer in African Americans, we have performed linkage analyses on members of 15 large high-risk families. Specifically, these families were recruited from Louisiana and represent a uniquely admixed African American population exclusive to Southern Louisiana. In addition to geographical constraints, these families were clinically homogeneous creating a well-characterized collection of large pedigrees. The families were genotyped with Illumina Infinium II SNP HumanLinkage-12 panel and extensive demographic and clinical information was documented from the hospital pathological reports and family interviews. RESULTS We identified two novel regions, 12q24 and 2p16, with suggestive evidence of linkage under the dominant model of inheritance. CONCLUSIONS This is the first time that chromosome 12q24 (HLOD = 2.21) and 2p16 (HLOD = 1.97) has been shown to be associated with prostate cancer in high-risk African American families. These results provide insight to prostate cancer in an exceptional, well-characterized African American population, and illustrate the significance of utilizing large unique, but homogenous pedigrees.
Collapse
Affiliation(s)
- Elisa M Ledet
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
18
|
Isaacs WB. Inherited susceptibility for aggressive prostate cancer. Asian J Androl 2012; 14:415-8. [PMID: 22543676 PMCID: PMC3568760 DOI: 10.1038/aja.2011.146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 03/30/2012] [Accepted: 03/30/2012] [Indexed: 01/17/2023] Open
Abstract
Whether or not there is inherited basis for prostate cancer aggressiveness is not clear, but advances in DNA analysis should provide an answer to this question in the very near future.
Collapse
Affiliation(s)
- William B Isaacs
- Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA.
| |
Collapse
|
19
|
Schumacher FR, Berndt SI, Siddiq A, Jacobs KB, Wang Z, Lindstrom S, Stevens VL, Chen C, Mondul AM, Travis RC, Stram DO, Eeles RA, Easton DF, Giles G, Hopper JL, Neal DE, Hamdy FC, Donovan JL, Muir K, Al Olama AA, Kote-Jarai Z, Guy M, Severi G, Grönberg H, Isaacs WB, Karlsson R, Wiklund F, Xu J, Allen NE, Andriole GL, Barricarte A, Boeing H, Bas Bueno-de-Mesquita H, Crawford ED, Diver WR, Gonzalez CA, Gaziano JM, Giovannucci EL, Johansson M, Le Marchand L, Ma J, Sieri S, Stattin P, Stampfer MJ, Tjonneland A, Vineis P, Virtamo J, Vogel U, Weinstein SJ, Yeager M, Thun MJ, Kolonel LN, Henderson BE, Albanes D, Hayes RB, Spencer Feigelson H, Riboli E, Hunter DJ, Chanock SJ, Haiman CA, Kraft P. Genome-wide association study identifies new prostate cancer susceptibility loci. Hum Mol Genet 2011; 20:3867-75. [PMID: 21743057 PMCID: PMC3168287 DOI: 10.1093/hmg/ddr295] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 06/18/2011] [Accepted: 06/27/2011] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PrCa) is the most common non-skin cancer diagnosed among males in developed countries and the second leading cause of cancer mortality, yet little is known regarding its etiology and factors that influence clinical outcome. Genome-wide association studies (GWAS) of PrCa have identified at least 30 distinct loci associated with small differences in risk. We conducted a GWAS in 2782 advanced PrCa cases (Gleason grade ≥ 8 or tumor stage C/D) and 4458 controls with 571 243 single nucleotide polymorphisms (SNPs). Based on in silico replication of 4679 SNPs (Stage 1, P < 0.02) in two published GWAS with 7358 PrCa cases and 6732 controls, we identified a new susceptibility locus associated with overall PrCa risk at 2q37.3 (rs2292884, P= 4.3 × 10(-8)). We also confirmed a locus suggested by an earlier GWAS at 12q13 (rs902774, P= 8.6 × 10(-9)). The estimated per-allele odds ratios for these loci (1.14 for rs2292884 and 1.17 for rs902774) did not differ between advanced and non-advanced PrCa (case-only test for heterogeneity P= 0.72 and P= 0.61, respectively). Further studies will be needed to assess whether these or other loci are differentially associated with PrCa subtypes.
Collapse
Affiliation(s)
- Fredrick R. Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Kevin B. Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Core Genotyping Facility, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
- Bioinformed Consulting Services, Gaithersburg, MD, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Core Genotyping Facility, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Sara Lindstrom
- Program in Molecular and Genetic Epidemiology
- Department of Epidemiology and
| | | | - Constance Chen
- Program in Molecular and Genetic Epidemiology
- Department of Epidemiology and
| | - Alison M. Mondul
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ruth C. Travis
- Cancer Epidemiology Unit
- Nuffield Department of Clinical Medicine and
| | - Daniel O. Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology
- Department of Public Health
- Department of Primary Care and
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Graham Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Victoria, Australia
| | - John L. Hopper
- Centre for Molecular, Environmental, Genetic and Analytic (MEGA) Epidemiology, Melbourne School of Population Health, The University of Melbourne, Melbourne, Australia
| | - David E. Neal
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Freddie C. Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Jenny L. Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Kenneth Muir
- Health Sciences Research Institute, University of Warwick, Coventry, UK
| | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology
- Department of Public Health
- Department of Primary Care and
| | | | - Michelle Guy
- Oncogenetics Team, The Institute of Cancer Research, Sutton, UK
| | - Gianluca Severi
- Cancer Epidemiology Centre, Cancer Council Victoria, Victoria, Australia
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - William B. Isaacs
- The Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jianfeng Xu
- Centers for Cancer Genomics and Center for Human Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Naomi E. Allen
- Cancer Epidemiology Unit
- Nuffield Department of Clinical Medicine and
| | - Gerald L. Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St Louis, MO, USA
| | | | - Heiner Boeing
- Department of Epidemiology, Deutsches Institut für Ernährungsforschung, Potsdam-Rehbrücke, Germany
| | | | | | - W. Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Carlos A. Gonzalez
- Unit of Nutrition, Environment and Cancer, Catalan Institute of Oncology (ICO-IDIBELL-RETICC RD06/0020), L'Hospitalet de Llobregat, Barcelona, Spain
| | - J. Michael Gaziano
- Division of Aging and
- Massachusetts Veterans Epidemiology Research and Information Center/VA Cooperative Studies Programs, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Edward L. Giovannucci
- Department of Epidemiology and
- Department of Nutrition, Harvard School of Public Health, Boston 02115, MA, USA
| | - Mattias Johansson
- International Agency for Research on Cancer (IARC), Lyon, France
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Loic Le Marchand
- Cancer Research Center of Hawaii, University of Hawaii, Honolulu, HI, USA
| | - Jing Ma
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Channing Laboratory, Boston, MA, USA
| | - Sabina Sieri
- Nutritional Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pär Stattin
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Meir J. Stampfer
- Department of Epidemiology and
- Department of Nutrition, Harvard School of Public Health, Boston 02115, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Channing Laboratory, Boston, MA, USA
| | - Anne Tjonneland
- Institute of Cancer Epidemiology, Danish Cancer Society, Copenhagen, Denmark
| | - Paolo Vineis
- MRC-HPA Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
- National Food Institute, Technical University of Denmark, Soborg, Denmark
| | - Stephanie J. Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Michael J. Thun
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | | | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Richard B. Hayes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Division of Epidemiology, Department of Environmental Medicine, New York University Langone Medical Center, NYU Cancer Institute, New York, NY, USA and
| | | | - Elio Riboli
- Department of Epidemiology and Biostatistics and
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology
- Department of Epidemiology and
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology
- Department of Epidemiology and
| |
Collapse
|
20
|
Liu X, Cheng I, Plummer SJ, Suarez B, Casey G, Catalona WJ, Witte JS. Fine-mapping of prostate cancer aggressiveness loci on chromosome 7q22-35. Prostate 2011; 71:682-9. [PMID: 20945404 PMCID: PMC3027848 DOI: 10.1002/pros.21284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 08/31/2010] [Indexed: 01/10/2023]
Abstract
BACKGROUND Deciphering the genetic basis of prostate cancer aggressiveness could provide valuable information for the screening and treatment of this common but complex disease. We previously detected linkage between a broad region on chromosome 7q22-35 and Gleason score-a strong predictor of prostate cancer aggressiveness. To further clarify this finding and focus on the potentially causative gene, we undertook a fine-mapping study across the 7q22-35 region. METHODS Our study population encompassed 698 siblings diagnosed with prostate cancer. 3,072 single nucleotide polymorphisms (SNPs) spanning the chromosome 7q22-35 region were genotyped using the Illumina GoldenGate assay. The impact of SNPs on Gleason scores were evaluated using affected sibling pair linkage and family-based association tests. RESULTS We confirmed the previous linkage signal and narrowed the 7q22-35 prostate cancer aggressiveness locus to a 370 kb region. Centered under the linkage peak is the gene KLRG2 (killer cell lectin-like receptor subfamily G, member 2). Association tests indicated that the potentially functional non-synonymous SNP rs17160911 in KLRG2 was significantly associated with Gleason score (P = 0.0007). CONCLUSIONS These findings suggest that genetic variants in the gene KLRG2 may affect Gleason score at diagnosis and hence the aggressiveness of prostate cancer.
Collapse
Affiliation(s)
- Xin Liu
- Mary Ann and J. Milburn Smith Child Health Research Program, Department of Pediatrics, Northwestern University Feinberg School of Medicine and Children's Memorial Hospital and Children's Memorial Research Center, Chicago, IL, USA
| | - Iona Cheng
- Epidemiology Program, Cancer Research Center of Hawai`i, University of Hawai`i, Honolulu, HI 96813, USA
| | - Sarah J Plummer
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Brian Suarez
- Department of Genetics, Washington University, 660 South Euclid, St. Louis, Missouri 63110, USA
| | - Graham Casey
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - William J. Catalona
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John S. Witte
- Departments of Epidemiology & Biostatistics and Urology, Institute for Human Genetics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158-9001, USA
| |
Collapse
|
21
|
Penney KL, Pyne S, Schumacher FR, Sinnott JA, Mucci LA, Kraft PL, Ma J, Oh WK, Kurth T, Kantoff PW, Giovannucci EL, Stampfer MJ, Hunter DJ, Freedman ML. Genome-wide association study of prostate cancer mortality. Cancer Epidemiol Biomarkers Prev 2010; 19:2869-76. [PMID: 20978177 DOI: 10.1158/1055-9965.epi-10-0601] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND A pressing clinical issue in prostate cancer is to distinguish which men will have an indolent or aggressive course of disease. Clinical variables such as Gleason grade and stage are useful predictors of lethal cancer; however, the low predictive values of the common Gleason scores, changes in grading over time, and earlier diagnosis of patients due to screening limits their clinical utility. Identifying genetic variants associated with lethal prostate cancer could inform clinical decision making. METHODS We conducted a genome-wide association study, comparing lethal prostate cancer cases to cases surviving at least 10 years beyond their initial diagnosis. Genotyping was done with the Affymetrix 5.0 chip [∼500,000 single nucleotide polymorphisms (SNP) and 1,483 copy number variants (CNV)] on DNA from participants in the Physicians' Health Study and Health Professionals Follow-up Study (196 lethal cases, 368 long-term survivors). After excluding SNPs and individuals based on quality control criteria, logistic regression assuming an additive model was done using the PLINK software. RESULTS No SNP reached genome-wide significance (P ≤ 1 × 10(-7)); however, three independent SNPs had P < 1 × 10(-5). One top-ranked SNP replicated (P = 0.05) in an independent follow-up study. Although no CNV had genome-wide significance, 14 CNVs showed nominal association with prostate cancer mortality (P < 0.05). CONCLUSIONS No variants were significantly associated at a genome-wide level with prostate cancer mortality. Common genetic determinants of lethal prostate cancer are likely to have odds ratios <2.0. IMPACT Genetic markers identified could provide biological insight to improve therapy for men with potentially fatal cancer. Larger studies are necessary to detect the genetic causes of prostate cancer mortality.
Collapse
Affiliation(s)
- Kathryn L Penney
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Christensen GB, Baffoe-Bonnie AB, George A, Powell I, Bailey-Wilson JE, Carpten JD, Giles GG, Hopper JL, Severi G, English DR, Foulkes WD, Maehle L, Moller P, Eeles R, Easton D, Badzioch MD, Whittemore AS, Oakley-Girvan I, Hsieh CL, Dimitrov L, Xu J, Stanford JL, Johanneson B, Deutsch K, McIntosh L, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Isaacs WB, Thibodeau SN, McDonnell SK, Hebbring S, Schaid DJ, Lange EM, Cooney KA, Tammela TLJ, Schleutker J, Paiss T, Maier C, Grönberg H, Wiklund F, Emanuelsson M, Farnham JM, Cannon-Albright LA, Camp NJ. Genome-wide linkage analysis of 1,233 prostate cancer pedigrees from the International Consortium for Prostate Cancer Genetics using novel sumLINK and sumLOD analyses. Prostate 2010; 70:735-44. [PMID: 20333727 PMCID: PMC3428045 DOI: 10.1002/pros.21106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Prostate cancer (PC) is generally believed to have a strong inherited component, but the search for susceptibility genes has been hindered by the effects of genetic heterogeneity. The recently developed sumLINK and sumLOD statistics are powerful tools for linkage analysis in the presence of heterogeneity. METHODS We performed a secondary analysis of 1,233 PC pedigrees from the International Consortium for Prostate Cancer Genetics (ICPCG) using two novel statistics, the sumLINK and sumLOD. For both statistics, dominant and recessive genetic models were considered. False discovery rate (FDR) analysis was conducted to assess the effects of multiple testing. RESULTS Our analysis identified significant linkage evidence at chromosome 22q12, confirming previous findings by the initial conventional analyses of the same ICPCG data. Twelve other regions were identified with genome-wide suggestive evidence for linkage. Seven regions (1q23, 5q11, 5q35, 6p21, 8q12, 11q13, 20p11-q11) are near loci previously identified in the initial ICPCG pooled data analysis or the subset of aggressive PC pedigrees. Three other regions (1p12, 8p23, 19q13) confirm loci reported by others, and two (2p24, 6q27) are novel susceptibility loci. FDR testing indicates that over 70% of these results are likely true positive findings. Statistical recombinant mapping narrowed regions to an average of 9 cM. CONCLUSIONS Our results represent genomic regions with the greatest consistency of positive linkage evidence across a very large collection of high-risk PC pedigrees using new statistical tests that deal powerfully with heterogeneity. These regions are excellent candidates for further study to identify PC predisposition genes.
Collapse
Affiliation(s)
- G Bryce Christensen
- University of Utah ICPCG Group and Division of Genetic Epidemiology, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fitzgerald LM, McDonnell SK, Carlson EE, Langeberg W, McIntosh LM, Deutsch K, Ostrander EA, Schaid DJ, Stanford JL. Genome-wide linkage analyses of hereditary prostate cancer families with colon cancer provide further evidence for a susceptibility locus on 15q11-q14. Eur J Hum Genet 2010; 18:1141-7. [PMID: 20407467 DOI: 10.1038/ejhg.2010.49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The search for susceptibility loci in hereditary prostate cancer (HPC) is challenging because of locus and disease heterogeneity. One approach to reduce disease heterogeneity is to stratify families on the basis of the occurrence of multiple cancer types. This method may increase the power for detecting susceptibility loci, including those with pleiotropic effects. We have completed a genome-wide SNP linkage analysis of 96 HPC families, each of which has one or more first-degree relatives with colon cancer (CCa), and further analyzed the subset of families with two or more CCa cases (n = 27). When only a prostate cancer (PCa) phenotype was considered to be affected, we observed suggestive evidence for linkage (LOD ≥1.86) at 15q14, 18q21 and 19q13 in all families, and at 1p32 and 15q11-q14 in families with two or more CCa cases. When both the PCa and CCa phenotypes were considered affected, suggestive evidence for linkage was observed at 11q25, 15q14 and 18q21 in all families, and at 1q31, 11q14 and 15q11-14 in families with two or more CCa cases. The strongest linkage signal was identified at 15q14 when both PCa and CCa phenotypes were considered to be affected in families with two or more CCa cases (recessive HLOD = 3.88). These results provide further support for the presence of HPC susceptibility loci on chromosomes 11q14, 15q11-q14 and 19q13 and highlight loci at 1q31, 11q, 15q11-14 and 18q21 as having possible pleiotropic effects. This study shows the benefit of using a comprehensive family cancer history to create more genetically homogenous subsets of HPC families for linkage analyses.
Collapse
Affiliation(s)
- Liesel M Fitzgerald
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
MicroRNA-330 acts as tumor suppressor and induces apoptosis of prostate cancer cells through E2F1-mediated suppression of Akt phosphorylation. Oncogene 2009; 28:3360-70. [PMID: 19597470 DOI: 10.1038/onc.2009.192] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) make up a novel class of gene regulators; they function as oncogenes or tumor suppressors by targeting tumor-suppressor genes or oncogenes. A recent study that analysed a large number of human cancer cell lines showed that miR-330 is a potential tumor-suppressor gene. However, the function and molecular mechanism of miR-330 in determining the aggressiveness of human prostate cancer has not been studied. Here, we show that miR-330 is significantly lower expressed in human prostate cancer cell lines than in nontumorigenic prostate epithelial cells. Bioinformatics analyses reveal a conserved target site for miR-330 in the 3'-untranslated region (UTR) of E2F1 at nucleotides 1018-1024. MiR-330 significantly suppressed the activity of a luciferase reporter containing the E2F1-3'-UTR in the cells. This activity could be abolished with the transfection of anti-miR-330 or mutated E2F1-3'-UTR. In addition, the expression level of miR-330 and E2F1 was inversely correlated in cell lines and prostate cancer specimens. After overexpressing of miR-330 in PC-3 cells, cell growth was suppressed by reducing E2F1-mediated Akt phosphorylation and thereby inducing apoptosis. Collectively, this is the first study to show that E2F1 is negatively regulated by miR-330 and also show that miR-330 induces apoptosis in prostate cancer cells through E2F1-mediated suppression of Akt phosphorylation.
Collapse
|
25
|
Stanford JL, FitzGerald LM, McDonnell SK, Carlson EE, McIntosh LM, Deutsch K, Hood L, Ostrander EA, Schaid DJ. Dense genome-wide SNP linkage scan in 301 hereditary prostate cancer families identifies multiple regions with suggestive evidence for linkage. Hum Mol Genet 2009; 18:1839-48. [PMID: 19251732 DOI: 10.1093/hmg/ddp100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The search for susceptibility loci in hereditary prostate cancer (HPC) has proven challenging due to genetic and disease heterogeneity. Multiple risk loci have been identified to date, however few loci have been replicated across independent linkage studies. In addition, most previous analyses have been hampered by the relatively poor information content provided by microsatellite scans. To overcome these issues, we have performed linkage analyses on members of 301 HPC families genotyped using the Illumina SNP linkage panel IVb. The information content for this panel, averaged over all pedigrees and all chromosomes, was 86% (range 83-87% over chromosomes). Analyses were also stratified on families according to disease aggressiveness, age at diagnosis and number of affected individuals to achieve more genetically homogeneous subsets. Suggestive evidence for linkage was identified at 7q21 (HLOD = 1.87), 8q22 (KCLOD = 1.88) and 15q13-q14 (HLOD = 1.99) in 289 Caucasian families, and nominal evidence for linkage was identified at 2q24 (LOD = 1.73) in 12 African American families. Analysis of more aggressive prostate cancer phenotypes provided evidence for linkage to 11q25 (KCLOD = 2.02), 15q26 (HLOD = 1.99) and 17p12 (HLOD = 2.13). Subset analyses according to age at diagnosis and number of affected individuals also identified several regions with suggestive evidence for linkage, including a KCLOD of 2.82 at 15q13-q14 in 128 Caucasian families with younger ages at diagnosis. The results presented here provide further evidence for a prostate cancer susceptibility locus on chromosome 15q and demonstrate the power of utilizing high information content SNP scans in combination with homogenous collections of large prostate cancer pedigrees.
Collapse
Affiliation(s)
- Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gho JWM, Ip WK, Chan KYY, Law PTY, Lai PBS, Wong N. Re-expression of transcription factor ATF5 in hepatocellular carcinoma induces G2-M arrest. Cancer Res 2008; 68:6743-51. [PMID: 18701499 DOI: 10.1158/0008-5472.can-07-6469] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transcription factors represent an important class of genes that play key roles in controlling cellular proliferation, cell cycle modulation, and attractive targets for cancer therapy. Here, we report on the novel finding of common ATF5 down-regulations in hepatocellular carcinoma (HCC), a highly malignant tumor with a dismal clinical course. Array-based mapping in HCC highlighted a high and consistent incidence of transcription factor ATF5 repressions on regional chr.19q13. By quantitative reverse transcription-PCR, profound down-regulations of ATF5 were further suggested in 78% of HCC tumors (60 of 77 cases) compared to their adjacent nontumoral liver (P = 0.0004). Restoration of ATF5 expression in 3 nonexpressing HCC cell lines demonstrated a consistent growth inhibitory effect (P < 0.029) but minimal induction on cellular apoptosis. Subsequent flow cytometric investigations revealed a G(2)-M cell cycle arrest in HCC cells that were ectopically transfected with ATF5 (P < 0.002). The differential expressed genes from the functional effects of ATF5 were examined by array profiling. Over a hundred genes were identified, among which ID1 contains the ATF/CREB target binding sequences within its promoter region. An inverse relationship between ATF5 expressions with ID1 transcriptions was verified in HCC (P = 0.019), and a direct interaction of ATF5 on the promoter of ID1 was further demonstrated from electromobility shift assay. Examination of causal events underlying the silencing of ATF5 in HCC suggested copy number losses, promoter hypermethylation, histone deacetylation, and DNA mutations to be the likely inactivating mechanisms. In conclusion, our finding supports a tumor suppressive role for ATF5 in HCC, and highlighted ID1 as a potential downstream target.
Collapse
Affiliation(s)
- Jennifer W-M Gho
- Li Ka-Shing Institute of Health Sciences, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
27
|
Ostrander EA, Johannesson B. Prostate cancer susceptibility loci: finding the genes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:179-90. [PMID: 18497042 DOI: 10.1007/978-0-387-69080-3_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Studies to date suggest that PC is a genetically very heterogeneous disease. High-risk families, in which multiple men are affected likely, reflect the contributions of a number of genes, some that are rare and highly penetrant, while others are more common and weakly penetrant. In this review, we have discussed only the first type of loci, and found that the identification of such genomic regions is a formidable problem. Replication between seemingly similar data sets is weak, likely reflecting the older age of onset associated with the disease, the inability to collect affected individuals from more than two generations in a family, and the variation seen in disease presentation, in addition to the underlying locus heterogeneity. Indeed, the definition of PC is ever changing, as diagnostic criteria and tools for pinpointing early lesions improve. Are we making progress? Clearly the answer is yes. The ability to divide large data sets into homogenous subset of families likely to share common genetic under-pinnings has improved power to identify loci and reproducibility between loci is now more common. Indeed, several groups report linkage to loci on chromosomes 1, 17, 19, and 22. Key to our continued success is our ever increasing ability to understand the disease. Identifying the subset of men who are likely to get clinically significant disease is the goal of genetic studies like these, and identifying the underlying loci is the key for developing diagnostics. The willingness of the community to work together has been an important factor in the successes the community has enjoyed to date, and will likely be as important as we move forward to untangle the genetics of this complex and common disorder.
Collapse
Affiliation(s)
- Elanie A Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
28
|
Cunningham JM, Hebbring SJ, McDonnell SK, Cicek MS, Christensen GB, Wang L, Jacobsen SJ, Cerhan JR, Blute ML, Schaid DJ, Thibodeau SN. Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomarkers Prev 2007; 16:969-78. [PMID: 17507624 DOI: 10.1158/1055-9965.epi-06-0767] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Previous studies suggest that enzymes involved in the androgen metabolic pathway are susceptibility factors for prostate cancer. Estrogen metabolites functioning as genotoxins have also been proposed as risk factors. In this study, we systematically tested the hypothesis that common genetic variations for those enzymes involved in the androgen and estrogen metabolic pathways increase risk for sporadic and familial prostate cancer. From these two pathways, 46 polymorphisms (34 single nucleotide polymorphisms, 10 short tandem repeat polymorphisms, and 2 null alleles) in 25 genes were tested for possible associations. Those genes tested included PRL, LHB, CYP11A1, HSD3B1, HSD3B2, HSD17B2, CYP17, SRD5A2, AKR1C3, UGT2B15, AR, SHBG, and KLK3 from the androgen pathway and CYP19, HSD17B1, CYP1A1, CYP1A2, CYP1B1, COMT, GSTP1, GSTT1, GSTM1, NQO1, ESR1, and ESR2 from the estrogen pathway. A case-control study design was used with two sets of cases: familial cases with a strong prostate cancer family history (n = 438 from 178 families) and sporadic cases with a negative prostate cancer family history (n = 499). The controls (n = 493) were derived from a population-based collection. Our results provide suggestive findings for an association with either familial or sporadic prostate cancer with polymorphisms in four genes: AKR1C3, HSD17B1, NQO1, and GSTT1. Additional suggestive findings for an association with clinical variables (disease stage, grade, and/or node status) were observed for single nucleotide polymorphisms in eight genes: HSD3B2, SRD5A2, SHBG, ESR1, CYP1A1, CYP1B1, GSTT1, and NQO1. However, none of the findings were statistically significant after appropriate corrections for multiple comparisons. Given that the point estimates for the odds ratio for each of these polymorphisms are <2.0, much larger sample sizes will be required for confirmation.
Collapse
Affiliation(s)
- Julie M Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Corson TW, Gallie BL. One hit, two hits, three hits, more? Genomic changes in the development of retinoblastoma. Genes Chromosomes Cancer 2007; 46:617-34. [PMID: 17437278 DOI: 10.1002/gcc.20457] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The childhood eye cancer retinoblastoma is initiated by the loss of both alleles of the prototypic tumor suppressor gene, RB1. However, a large number of cytogenetic and comparative genomic hybridization (CGH) studies have shown that these M1 and M2 mutational events--although necessary for initiation--are not the only genomic changes in retinoblastoma. Some of these subsequent changes, which we have termed M3 to Mn, are likely crucial for tumor progression not only in retinoblastoma but also in other cancers. Moreover, genes showing genomic change in cancer are more stable markers and, therefore, possible therapeutic targets than genes simply differentially expressed. In this review, we provide the first comprehensive summary of the genomic evidence implicating gain of 1q, 2p, 6p, and 13q, and loss of 16q in retinoblastoma oncogenesis, including karyotype, CGH, and microarray CGH data. We discuss the search for candidate oncogenes and tumor suppressor genes within these regions, including the candidates (KIF14, MDM4, MYCN, E2F3, DEK, CDH11, and others), plus associations between genomic changes and clinical parameters. We also review studies of other regions of the retinoblastoma genome, the epigenetic changes of aberrant methylation of MGMT, RASSF1A, CASP8, and MLH1, and the roles microRNAs might play in this cancer. Although many candidate genes have yet to be functionally validated in retinoblastoma, work in this field lays out a molecular cytogenetic pathway of retinoblastoma development. Candidate cancer genes carry diagnostic, prognostic, and therapeutic implications beyond retinoblastoma.
Collapse
Affiliation(s)
- Timothy W Corson
- Division of Applied Molecular Oncology, Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | | |
Collapse
|
30
|
Pal P, Xi H, Sun G, Kaushal R, Meeks JJ, Thaxton CS, Guha S, Jin CH, Suarez BK, Catalona WJ, Deka R. Tagging SNPs in the kallikrein genes 3 and 2 on 19q13 and their associations with prostate cancer in men of European origin. Hum Genet 2007; 122:251-9. [PMID: 17593395 DOI: 10.1007/s00439-007-0394-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
Two of the classical kallikrein genes KLK3 and KLK2 on 19q13.4 are plausible candidates in prostate cancer susceptibility. They are expressed almost exclusively in prostate tissue. We have performed a comprehensive analysis of association of variants in these two genes with prostate cancer among men of European descent using a tagging SNP approach. Thirteen SNPs selected from the HapMap database were analyzed in a sample of 596 histologically verified prostate cancer cases and 567 ethnically matched controls. Five SNPs showed significant association at single marker level. Linkage disequilibrium (LD) analysis revealed four LD blocks. We performed a haplotype analysis within each LD block. A major haplotype in block 1 that contains the first two significantly associated SNPs was significantly underrepresented in the prostate cancer cases; a second haplotype in block 3 also showed significant frequency differences between cases and controls. Four of the studied SNPs show positive associations with serum PSA levels. A structure analysis revealed no population stratification in our samples that could have confounded the association results. These findings suggest a plausible role of kallikrein gene variants in the etiology of prostate cancer among men of European ancestry.
Collapse
Affiliation(s)
- Prodipto Pal
- Department of Environmental Health, Center for Genome Information, University of Cincinnati Medical Center, OH 45267-0056, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ostrander EA, Kwon EM, Stanford JL. Genetic susceptibility to aggressive prostate cancer. Cancer Epidemiol Biomarkers Prev 2007; 15:1761-4. [PMID: 17035380 DOI: 10.1158/1055-9965.epi-06-0730] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Elaine A Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, 50 South Drive, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
32
|
Suuriniemi M, Agalliu I, Schaid DJ, Johanneson B, McDonnell SK, Iwasaki L, Stanford JL, Ostrander EA. Confirmation of a positive association between prostate cancer risk and a locus at chromosome 8q24. Cancer Epidemiol Biomarkers Prev 2007; 16:809-14. [PMID: 17416775 DOI: 10.1158/1055-9965.epi-06-1049] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Family-based linkage studies, association studies, and studies of tumors have highlighted human chromosome 8q as a genomic region of interest for prostate cancer susceptibility loci. Recently, a locus at 8q24, characterized by both a single nucleotide polymorphism (SNP) and a microsatellite marker, was shown to be associated with prostate cancer risk in Icelandic, Swedish, and U.S. samples. Although the data were provocative, the U.S. samples were not population based, which precludes assessment of the potential contribution of this locus to prostate cancer incidence in the United States. METHODS We analyzed both markers in a population-based, case-control study of middle-aged men from King County, Washington. RESULTS Overall, there was a significant positive association between the A allele of the SNP rs1447295 and prostate cancer risk [odds ratio, 1.4; 95% confidence interval (95% CI), 1.1-2.0] but no significant association with the microsatellite DG8S737. However, significant associations were observed for both markers in men with high Gleason scores. Adjusting for age, first-degree family history of prostate cancer, and prostate cancer screening history, the adjusted odds ratios were 1.4 (95% CI, 1.1-1.8) for the A allele of the SNP and 1.9 (95% CI, 1.2-2.8) for the -10 allele of the microsatellite. CONCLUSIONS These data suggest that the locus on chromosome 8q24 harbors a genetic variant associated with prostate cancer and that the microsatellite marker is a stronger risk factor for aggressive prostate cancers defined by poorly differentiated tumor morphology.
Collapse
Affiliation(s)
- Miia Suuriniemi
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Schaid DJ, Stanford JL, McDonnell SK, Suuriniemi M, McIntosh L, Karyadi DM, Carlson EE, Deutsch K, Janer M, Hood L, Ostrander EA. Genome-wide linkage scan of prostate cancer Gleason score and confirmation of chromosome 19q. Hum Genet 2007; 121:729-35. [PMID: 17486369 DOI: 10.1007/s00439-007-0368-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 04/12/2007] [Indexed: 11/26/2022]
Abstract
Despite evidence that prostate cancer has a genetic etiology, it has been extremely difficult to confirm genetic linkage results across studies, emphasizing the large extent of genetic heterogeneity associated with this disease. Because prostate cancer is common--approximately one in six men will be diagnosed with prostate cancer in their life--genetic linkage studies are likely plagued by phenocopies (i.e., men with prostate cancer due to environmental or lifestyle factors), weakly penetrant alleles, or a combination of both, making it difficult to replicate linkage findings. One way to account for heterogeneous causes is to use clinical information that is related to the aggressiveness of disease as an endpoint for linkage analyses. Gleason grade is a measure of prostate tumor differentiation, with higher grades associated with more aggressive disease. This semi-quantitative score has been used as a quantitative trait for linkage analysis in several prior studies. Our aim was to determine if prior linkage reports of Gleason grade to specific loci could be replicated, and to ascertain if new regions of linkage could be found. Gleason scores were available for 391 affected sib pairs from 183 hereditary prostate cancer pedigrees as part of the PROGRESS study. Analyzing Gleason score as a quantitative trait, and using microsatellite markers, suggestive evidence for linkage (P-value <or= 0.001) was found on chromosomes 19q and 5q, with P-values <or= 0.01 observed on chromosomes 3q, 7q, and 16q. Our results confirm reports of Gleason score linkage to chromosome 19q and suggest new loci for further investigation.
Collapse
|
34
|
Christensen GB, Camp NJ, Farnham JM, Cannon-Albright LA. Genome-wide linkage analysis for aggressive prostate cancer in Utah high-risk pedigrees. Prostate 2007; 67:605-13. [PMID: 17299800 DOI: 10.1002/pros.20554] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND It has been proposed that studying alternative phenotypes, such as tumor aggressiveness, may be a solution for overcoming the apparent heterogeneity that has hindered the identification of prostate cancer (PC) genes. We present the results of a genome-scan for predisposition to aggressive PC using the Utah high-risk pedigree resource. METHODS We identified 259 subjects with aggressive PC in 57 extended and nuclear families. Parametric and non-parametric multipoint linkage statistics were calculated for a genome-wide set of 401 microsatellite markers using the MCLINK software package. Stratification analyses by the number of affected subjects per pedigree (<5, >or=5) and the average age at diagnosis of affected subjects (<70 years, >or=70 years) were also performed. RESULTS No significant results were observed at the genome-wide level, but suggestive evidence for linkage was observed on chromosomes 9q (HLOD = 2.04) and 14q (HLOD = 2.08); several pedigrees showed individual evidence for linkage at each locus (LOD > 0.58). The subset of pedigrees with earlier age at onset demonstrated nominal linkage evidence on chromosomes 3q (HLOD = 1.79), 8q (HLOD = 1.67), and 20q (HLOD=1.82). The late-onset subset showed suggestive linkage on chromosome 6p (HLOD = 2.37) and the subset of pedigrees with fewer than five affected subjects showed suggestive linkage on chromosome 10p (HLOD = 1.99). CONCLUSIONS Linkage evidence observed on chromosomes 6p, 8q, and 20q support previously reported PC aggressiveness loci. While these results are encouraging, further research is necessary to identify the gene or genes responsible for PC aggressiveness and surmount the overarching problem of PC heterogeneity.
Collapse
Affiliation(s)
- G B Christensen
- Department of Biomedical Informatics, University of Utah School of Medicine, Utah, USA.
| | | | | | | |
Collapse
|
35
|
Wernert N, Kaminski A, Haddouti EM, Hahne JC. Tumor-stroma interactions of metastatic prostate cancer cell lines: analyses using microarrays. Methods Mol Biol 2007; 382:223-237. [PMID: 18220234 DOI: 10.1007/978-1-59745-304-2_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Tumor-stroma interactions are of great importance not only for the development and progression of primary prostate carcinoma but probably also for the establishment of metastasis. Fibroblasts are an important stromal cell type encountered by metastatic tumor cells at different sites. In previous investigations, we had found that media conditioned by three metastatic prostate cancer cell lines (LNCaP, PC-3, and DU-145) induced cultured nonprostatic fibroblasts to proliferate or to express matrix-metalloproteinase-1 considered important for tumor invasion. Fibroblast-conditioned media in turn stimulate proliferation of DU-145 cells and migration of PC-3 cells. Both tumor cells and fibroblasts secrete VEGF suggesting that not only metastatic but also stromal cells at metastatic sites contribute to the vascularization of metastasis necessary for continuous growth. In order to better understand the reciprocal tumor-stroma cross-talk in molecular terms we used the mRNA extracted from stimulated and unstimulated neoplastic and fibroblastic stromal cells for cDNA array hybridization using Affymetrix chips. The three prostate cell lines influenced the fibroblasts nearly in the same manner. In particular proteins involved in cell adhesion, cell-cell contact, and cell cycle regulation were downregulated in stimulated fibroblasts. In contrast, fibroblasts affected every prostate cancer cell line in different ways, which may be because of the different origin of the metastatic prostate cancer cell lines.
Collapse
|
36
|
Schaid DJ, McDonnell SK, Zarfas KE, Cunningham JM, Hebbring S, Thibodeau SN, Eeles RA, Easton DF, Foulkes WD, Simard J, Giles GG, Hopper JL, Mahle L, Moller P, Badzioch M, Bishop DT, Evans C, Edwards S, Meitz J, Bullock S, Hope Q, Guy M, Hsieh CL, Halpern J, Balise RR, Oakley-Girvan I, Whittemore AS, Xu J, Dimitrov L, Chang BL, Adams TS, Turner AR, Meyers DA, Friedrichsen DM, Deutsch K, Kolb S, Janer M, Hood L, Ostrander EA, Stanford JL, Ewing CM, Gielzak M, Isaacs SD, Walsh PC, Wiley KE, Isaacs WB, Lange EM, Ho LA, Beebe-Dimmer JL, Wood DP, Cooney KA, Seminara D, Ikonen T, Baffoe-Bonnie A, Fredriksson H, Matikainen MP, Tammela TLJ, Bailey-Wilson J, Schleutker J, Maier C, Herkommer K, Hoegel JJ, Vogel W, Paiss T, Wiklund F, Emanuelsson M, Stenman E, Jonsson BA, Grönberg H, Camp NJ, Farnham J, Cannon-Albright LA, Catalona WJ, Suarez BK, Roehl KA. Pooled genome linkage scan of aggressive prostate cancer: results from the International Consortium for Prostate Cancer Genetics. Hum Genet 2006; 120:471-85. [PMID: 16932970 DOI: 10.1007/s00439-006-0219-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 06/05/2006] [Indexed: 10/24/2022]
Abstract
While it is widely appreciated that prostate cancers vary substantially in their propensity to progress to a life-threatening stage, the molecular events responsible for this progression have not been identified. Understanding these molecular mechanisms could provide important prognostic information relevant to more effective clinical management of this heterogeneous cancer. Hence, through genetic linkage analyses, we examined the hypothesis that the tendency to develop aggressive prostate cancer may have an important genetic component. Starting with 1,233 familial prostate cancer families with genome scan data available from the International Consortium for Prostate Cancer Genetics, we selected those that had at least three members with the phenotype of clinically aggressive prostate cancer, as defined by either high tumor grade and/or stage, resulting in 166 pedigrees (13%). Genome-wide linkage data were then pooled to perform a combined linkage analysis for these families. Linkage signals reaching a suggestive level of significance were found on chromosomes 6p22.3 (LOD = 3.0), 11q14.1-14.3 (LOD = 2.4), and 20p11.21-q11.21 (LOD = 2.5). For chromosome 11, stronger evidence of linkage (LOD = 3.3) was observed among pedigrees with an average at diagnosis of 65 years or younger. Other chromosomes that showed evidence for heterogeneity in linkage across strata were chromosome 7, with the strongest linkage signal among pedigrees without male-to-male disease transmission (7q21.11, LOD = 4.1), and chromosome 21, with the strongest linkage signal among pedigrees that had African American ancestry (21q22.13-22.3; LOD = 3.2). Our findings suggest several regions that may contain genes which, when mutated, predispose men to develop a more aggressive prostate cancer phenotype. This provides a basis for attempts to identify these genes, with potential clinical utility for men with aggressive prostate cancer and their relatives.
Collapse
Affiliation(s)
- Daniel J Schaid
- Harwick 7, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Williams YN, Masuda M, Sakurai-Yageta M, Maruyama T, Shibuya M, Murakami Y. Cell adhesion and prostate tumor-suppressor activity of TSLL2/IGSF4C, an immunoglobulin superfamily molecule homologous to TSLC1/IGSF4. Oncogene 2006; 25:1446-53. [PMID: 16261159 DOI: 10.1038/sj.onc.1209192] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The TSLL2/IGSF4C encodes an immunoglobulin (Ig) superfamily molecule showing significant homology with a lung tumor suppressor, TSLC1. The TSLL2 protein of 55 kDa is mainly expressed in the kidney, bladder, and prostate in addition to the brain. Here, we report the biological significance of TSLL2 in the urinary tissues. An immunohistochemical study reveals that TSLL2 is expressed at the cell-cell attachment sites in the renal tubules, the transitional epithelia of the bladder, and the glandular epithelia of the prostate. Confocal microscopy analysis demonstrates that TSLL2 is localized in the lateral membranes in polarized Mardin-Darby canine kidney (MDCK) cells. TSLL2 forms homo-dimers and its overexpression induces aggregation of suspended MDCK cells in a Ca2+/Mg2+-independent manner, suggesting that it is involved in cell adhesion through homophilic trans-interaction. The TSLL2 gene is mapped on the chromosomal region 19q13.2, whose loss of heterozygosity has been frequently reported in prostate cancer. TSLL2 protein is lost in nine of nine primary prostate cancers and in a prostate cancer cell, PPC-1. Introduction of TSLL2 into PPC-1 strongly suppresses subcutaneous tumor formation in nude mice. These results suggest that TSLL2 is a new member of the Ig superfamily cell adhesion molecules and is a tumor-suppressor candidate in prostate cancer.
Collapse
Affiliation(s)
- Y N Williams
- Tumor Suppression and Functional Genomics Project, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Lange EM, Ho LA, Beebe-Dimmer JL, Wang Y, Gillanders EM, Trent JM, Lange LA, Wood DP, Cooney KA. Genome-wide linkage scan for prostate cancer susceptibility genes in men with aggressive disease: significant evidence for linkage at chromosome 15q12. Hum Genet 2006; 119:400-7. [PMID: 16508751 DOI: 10.1007/s00439-006-0149-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 01/23/2006] [Indexed: 01/02/2023]
Abstract
Epidemiological and twin studies have consistently demonstrated a strong genetic component to prostate cancer (PCa) susceptibility. To date, numerous linkage studies have been performed to identify chromosomal regions containing PCa susceptibility genes. Unfortunately, results from these studies have failed to form any obvious consensus regarding which regions are most likely to contain genes that may contribute to PCa predisposition. One plausible explanation for the difficulty in mapping susceptibility loci is the existence of considerable heterogeneity in the phenotype of PCa, with significant variation in clinical stage and grade of disease even among family members. To address this issue, we performed a genome-wide linkage scan on 71 informative families with two or more men with aggressive PCa. When only men with aggressive PCa were coded as affected, statistically significant evidence for linkage at chromosome 15q12 was detected (LOD=3.49; genome-wide p=0.005). Furthermore, the evidence for linkage increased when analyses were restricted to Caucasian-American pedigrees (n=65; LOD=4.05) and pedigrees with two confirmed aggressive cases (n=42, LOD=4.76). Interestingly, a 1-LOD support interval about our peak at 15q12 overlaps a region of suggestive linkage, 15q11, identified by a recent linkage study on 1,233 PCa families by the International Consortium for Prostate Cancer Genetics. Using a more rigid definition of PCa in linkage studies will result in a severe reduction in sample sizes available for study, but may ultimately prove to increase statistical power to detect susceptibility genes for this multigenic trait.
Collapse
Affiliation(s)
- Ethan M Lange
- Department of Genetics, University of North Carolina, 4300D MBRB, CB# 7264, 103 Mason Farm Road, Chapel Hill, NC 27599-7264, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Stanford JL, McDonnell SK, Friedrichsen DM, Carlson EE, Kolb S, Deutsch K, Janer M, Hood L, Ostrander EA, Schaid DJ. Prostate cancer and genetic susceptibility: a genome scan incorporating disease aggressiveness. Prostate 2006; 66:317-25. [PMID: 16245279 DOI: 10.1002/pros.20349] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Prostate cancer is a heterogeneous disease, both genetically and phenotypically. Linkage studies attempting to map genes for hereditary prostate cancer (HPC) have proved challenging, and one potential problem contributing to this challenge is the variability in disease phenotypes. METHODS We collected clinical data on 784 affected men with prostate cancer from 248 HPC families for whom a genomic screen was performed. Disease characteristics (i.e., Gleason score, stage, prostate-specific antigen (PSA)) were used to classify affected men into categories of clinically insignificant, moderate, or aggressive prostate cancer. To potentially enrich for a genetic etiology, we restricted linkage analyses to only men with aggressive disease, although we used genotype information from all family members; linkage analyses used both dominant and recessive models. In addition, subset analyses considered age at diagnosis, number of affected men per family and other stratifications to try to increase genetic homogeneity. RESULTS Several regions of interest (heterogeneity LOD score, HLOD>1.0) were identified in families (n=123) with >or=2 affecteds with aggressive prostate cancer. "Suggestive" linkage was observed at chromosome 22q11.1 (Dominant model HLOD=2.18) and the result was stronger (Dominant HLOD=2.75) in families with evidence of male-to-male transmission. A second region at 22q12.3-q13.1 was also highlighted (Recessive model HLOD=1.90) among men with aggressive disease, as was a region on chromosome 18. CONCLUSIONS These analyses suggest that using clinically defined phenotypes may be a useful approach for simplifying the locus heterogeneity problems that confound the search for prostate cancer susceptibility genes.
Collapse
Affiliation(s)
- Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, and Department of Epidemiology, School of Public Health and Community Medicine, University of Washington, Seattle, Washington 98109-1024, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Slager SL, Zarfas KE, Brown WM, Lange EM, McDonnell SK, Wojno KJ, Cooney KA. Genome-wide linkage scan for prostate cancer aggressiveness loci using families from the University of Michigan Prostate Cancer Genetics Project. Prostate 2006; 66:173-9. [PMID: 16173044 DOI: 10.1002/pros.20332] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Prostate cancer (PC) is a complex disease that displays variable disease outcome, ranging from a relatively indolent disease to forms that result in death from the disease. One measure of disease severity is the Gleason score. Using the Gleason score as a measure of tumor aggressiveness, several independent genome scans have reported evidence of linkage. As of yet, however, no genes have been implicated. METHODS We report an independent genome scan using the Gleason score as a quantitative trait. We genotyped 405 highly polymorphic microsatellite markers in 175 brother pairs from 103 families. RESULTS Our strongest evidence of linkage is to 6q23 at 137 cM (D6S292, P = 0.0009). Other interesting regions (P < 0.005) were on chromosome 1p13-q21 and on chromosome 5p13-q11. CONCLUSIONS Our results provide further evidence that tumor aggressiveness has a genetic component, and that this genetic component may be influenced by several independent genes.
Collapse
Affiliation(s)
- Susan L Slager
- Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Cicek MS, Liu X, Casey G, Witte JS. Role of Androgen Metabolism Genes CYP1B1, PSA/KLK3, and CYP11 in Prostate Cancer Risk and Aggressiveness. Cancer Epidemiol Biomarkers Prev 2005; 14:2173-7. [PMID: 16172228 DOI: 10.1158/1055-9965.epi-05-0215] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Candidate genes involved with androgen metabolism have been hypothesized to affect the risk of prostate cancer. To further investigate this, we evaluated the relationship between prostate cancer and multiple potentially functional polymorphisms in three genes involved in androgen metabolism: CYP1B1 (two single nucleotide polymorphisms: 355G/T and 4326C/G), prostate-specific antigen (PSA/KLK3 (three single nucleotide polymorphisms: -158A/G, -4643G/A, and -5412C/T), and CYP11alpha [(tttta)(n) repeat], using a moderately large (n = 918) sibling-based case-control population. When looking at all subjects combined, no association was observed between any polymorphism-or their haplotypes-and prostate cancer risk. However, among men with more aggressive prostate cancer, the CYP1B1 355G/T variant was positively associated with disease: carrying one or two T alleles gave odds ratios (OR) of 1.90 [95% confidence interval (95% CI), 1.09-3.31; P = 0.02] and 3.73 (95% CI, 1.39-10.0; P = 0.009), respectively. Similarly, carrying the CYP1B1 355T-4326C haplotype was positively associated with prostate cancer among men with high aggressive disease (P = 0.01). In addition, the PSA -158G/-158G genotype was positively associated with prostate cancer among men with less aggressive disease (OR, 2.71; 95% CI, 1.06-6.94; P = 0.04). Our findings suggest that CYP1B1 and PSA variants may affect the risk of prostate cancer and tumor aggressiveness.
Collapse
Affiliation(s)
- Mine S Cicek
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Lerner School of Medicine, Ohio, USA
| | | | | | | |
Collapse
|
42
|
Yang P, Kollmeyer TM, Buckner K, Bamlet W, Ballman KV, Jenkins RB. Polymorphisms in GLTSCR1 and ERCC2 are associated with the development of oligodendrogliomas. Cancer 2005; 103:2363-72. [PMID: 15834925 DOI: 10.1002/cncr.21028] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Deletions of 19q have been associated with gliomas, especially oligodendrogliomas. In addition, cases with oligodendrogliomas with the 19q deletion have been observed to have a better survival compared with cases without the 19q deletion. The authors have previously described a 150-kilobase minimal deletion region in gliomas that maps to 19q13.33 and contains 3 novel candidate genes (GLTSCR1, EHD2, and GLTSCR2). METHODS The authors performed an association study using 141 cases with gliomas (61 cases with astrocytomas, 40 cases with oligodendrogliomas, 40 cases with mixed oligoastrocytomas) and 108 general controls. They evaluated 7 single nucleotide polymorphisms (SNPs) in 6 genes within and nearby the minimal 19q deletion region (ERCC2, RAI, ASE-1, ERCC1, GLTSCR1, and LIG1). RESULTS The prevalence of a germline GLTSCR1-exon-1 T allele (SNP rs1035938) was 40% in cases with oligodendrogliomas compared with 27% in controls (P = 0.029), and the prevalence of an ERCC2-exon-22 T allele (SNP rs1052555) was 35% in cases with oligodendrogliomas compared with 18% in controls (P = 0.043). One high-risk and 1 low-risk haplotype were associated with oligodendroglioma development (P = 0.003 and 0.026, respectively). Cases with oligodendrogliomas with the 19q deletion had a significantly higher frequency of the GLTSCR1-exon-1 T allele compared with cases without the 19q deletion (P = 0.01). It was noteworthy that cases with gliomas who were homozygous for the GLTSCR1-exon-1 T allele had a significantly better survival: 77% and 68% survival at 2 and 5 years compared with 56% and 34% for other genotypes (P = 0.02, log-rank test). Multivariable analysis identified grade, age, and the GLTSCR1-exon-1 and ERCC2-exon-22 genotypes as independent predictors for survival. CONCLUSIONS These results suggested that alterations in GLTSCR1 (or a closely linked gene) were associated with the development and progression of oligodendroglioma.
Collapse
Affiliation(s)
- Ping Yang
- Department of Health Sciences Research, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Nexø BA, Vogel U, Overvad K, Raaschou-Nielsen O, Tjønneland A. Who gets cancer? Breast Cancer Res 2005. [PMCID: PMC4233520 DOI: 10.1186/bcr1099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
44
|
Schaid DJ, Chang BL. Description of the International Consortium For Prostate Cancer Genetics, and failure to replicate linkage of hereditary prostate cancer to 20q13. Prostate 2005; 63:276-90. [PMID: 15599943 DOI: 10.1002/pros.20198] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The International Consortium for Prostate Cancer Genetics (ICPCG) is an international collaborative effort to pool pedigrees with hereditary prostate cancer (PC) in order to replicate linkage findings for PC. A strength of the ICPCG is the large number of well-characterized pedigrees, allowing linkage analyses within large subsets. Given the heterogeneity and complexity of PC, the historical difficulties of synthesizing different studies reporting positive and negative linkage replication, and the use of different statistical analysis methods and different stratification criteria, the ICPCG provides a valuable resource to evaluate linkage for hereditary PC. To date, linkage of chromosome 20 (HPC20) to hereditary PC has been one of the strongest linkage signals, yet the efforts to replicate this linkage have been limited. This paper reports a linkage analysis of chromosome 20 markers for 1,234 pedigrees with multiple cases of PC ascertained through the ICPCG, and represents the most thorough attempt to confirm or refute linkage to chromosome 20. From the original 158 Mayo pedigrees in which linkage was detected, the maximum heterogeneity LOD (HLOD) score, under a recessive model, was 2.78. In contrast, for the 1,076 pedigrees not included in the original study, the maximum HLOD score (recessive model) was 0.06. Although, a few small linkage signals for chromosome 20 were found in various strata of this pooled analysis, this large study failed to replicate linkage to HPC20. This study illustrates the value of the ICPCG family collection to evaluate reported linkage signals and suggests that the HPC20 region does not make a major contribution to PC susceptibility.
Collapse
Affiliation(s)
- Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | |
Collapse
|
45
|
Abstract
Prostate cancer is a heterogeneous disease with multiple loci contributing to susceptibility. Traditionally, genome-wide scans using high-risk families have utilized stratification by number of affected individuals, family history of other cancers, or family age at diagnosis to improve genetic homogeneity. In addition to locus heterogeneity, for later onset diseases such as prostate cancer, a major limitation to mapping efforts is that key parental DNA samples are rarely available. The lack of available samples from upper generations reduces inheritance information, and as a result, the standard 10-cM genome scan does not provide full power to detect linkage. To increase the ability to find disease-associated loci, much denser genome-wide scans must be undertaken in multiple ethnic groups. In addition, new ways of defining homogenous subsets of families need to be developed.
Collapse
Affiliation(s)
- Elaine A Ostrander
- Division of Clinical Research1, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024, USA.
| | | | | |
Collapse
|
46
|
Edwards SM, Eeles RA. Unravelling the genetics of prostate cancer. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2004; 129C:65-73. [PMID: 15264274 DOI: 10.1002/ajmg.c.30027] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review describes what is currently known about the genetics of prostate cancer. Traditionally, the genetics of a suspected inherited cancer predisposition have generally been thought of in terms of a single, high-risk gene with a dominant mode of inheritance. Such a gene might be observed in families, as has been documented in familial breast cancer (BRCA1/2), familial colorectal cancer (HNPCC), retinoblastoma (RB1), and Wilms tumor (WT1). This review investigates the evidence for the existence, first of familial prostate cancer, and second, for the presence of such a high-risk gene in those families by epidemiological and experimental approaches. Another current area of interest in prostate cancer is the investigation of the contribution of common lower penetrance genes to the disease. This alternative approach has become popular, as it raises the issue of frequently seen genetic variations such as single nucleotide polymorphisms (SNPs) having relevance to the risk of developing the disease. Finally, this article will explore the way forward, with emphasis on worldwide collaboration from teams attempting to find the genes responsible for the disease and investment in new technologies that will aid in their discovery.
Collapse
Affiliation(s)
- Stephen M Edwards
- Translational Cancer Genetics Team, Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | | |
Collapse
|
47
|
Kommu S, Edwards S, Eeles R. The clinical genetics of prostate cancer. Hered Cancer Clin Pract 2004; 2:111-21. [PMID: 20233465 PMCID: PMC4392519 DOI: 10.1186/1897-4287-2-3-111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 07/27/2004] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer is the most common cancer in men and the second highest cause of cancer-related mortality in the U.K. A genetic component in predisposition to prostate cancer has been recognized for decades. One of the strongest epidemiological risk factors for prostate cancer is a positive family history. The hunt for the genes that predispose to prostate cancer in families has been the focus of many research groups worldwide for the past 10 years. Both epidemiological and twin studies support a role for genetic predisposition to prostate cancer. Familial cancer loci have been found, but the genes that cause familial prostate cancer remain largely elusive. Unravelling the genetics of prostate cancer is challenging and is likely to involve the analysis of numerous predisposition genes. Current evidence supports the hypothesis that excess familial risk of prostate cancer could be due to the inheritance of multiple moderate-risk genetic variants. Although research on hereditary prostate cancer has improved our knowledge of the genetic aetiology of the disease, a lot of questions still remain unanswered. This article explores the current evidence that there is a genetic component to the aetiology of prostate cancer and attempts to put into context the diverse findings that have been shown to be possibly associated with the development of hereditary prostate cancer. Linkage searches over the last decade are summarised. It explores issues as to why understanding the genetics of prostate cancer has been so difficult and why despite this, it is still a major focus of research. Finally, current and future management strategies of men with Hereditary Prostate Cancer (HPC) are discussed.
Collapse
Affiliation(s)
- Sashi Kommu
- Reader in Clinical Cancer Genetics, Translational Cancer Genetics Team, Institute of Cancer Research, Sutton, UK.
| | | | | |
Collapse
|
48
|
Wang Q, Rao S, Shen GQ, Li L, Moliterno DJ, Newby LK, Rogers WJ, Cannata R, Zirzow E, Elston RC, Topol EJ. Premature myocardial infarction novel susceptibility locus on chromosome 1P34-36 identified by genomewide linkage analysis. Am J Hum Genet 2004; 74:262-71. [PMID: 14732905 PMCID: PMC1181924 DOI: 10.1086/381560] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Accepted: 11/18/2003] [Indexed: 11/03/2022] Open
Abstract
The most frequent causes of death and disability in the Western world are atherosclerotic coronary artery disease (CAD) and acute myocardial infarction (MI). This common disease is thought to have a polygenic basis with a complex interaction with environmental factors. Here, we report results of a genomewide search for susceptibility genes for MI in a well-characterized U.S. cohort consisting of 1,613 individuals in 428 multiplex families with familial premature CAD and MI: 712 with MI, 974 with CAD, and average age of onset of 44.4+/-9.7 years. Genotyping was performed at the National Heart, Lung, and Blood Institute Mammalian Genotyping Facility through use of 408 markers that span the entire human genome every 10 cM. Linkage analysis was performed with the modified Haseman-Elston regression model through use of the SIBPAL program. Three genomewide scans were conducted: single-point, multipoint, and multipoint performed on of white pedigrees only (92% of the cohort). One novel significant susceptibility locus was detected for MI on chromosomal region 1p34-36, with a multipoint allele-sharing P value of <10(-12) (LOD=11.68). Validation by use of a permutation test yielded a pointwise empirical P value of.00011 at this locus, which corresponds to a genomewide significance of P<.05. For the less restrictive phenotype of CAD, no genetic locus was detected, suggesting that CAD and MI may not share all susceptibility genes. The present study thus identifies a novel genetic-susceptibility locus for MI and provides a framework for the ultimate cloning of a gene for the complex disease MI.
Collapse
Affiliation(s)
- Qing Wang
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Shaoqi Rao
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Gong-Qing Shen
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Lin Li
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - David J. Moliterno
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - L. Kristin Newby
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - William J. Rogers
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Ruth Cannata
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Erich Zirzow
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Robert C. Elston
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| | - Eric J. Topol
- Center for Cardiovascular Genetics, Departments of Cardiovascular Medicine and Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, and Departments of Epidemiology and Biostatistics, and Medicine, Cleveland Clinic College of Medicine of Case Western Reserve University, Cleveland; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; and Department of Medicine, The University of Alabama Medical Center, Birmingham
| |
Collapse
|
49
|
Bostwick DG, Burke HB, Djakiew D, Euling S, Ho SM, Landolph J, Morrison H, Sonawane B, Shifflett T, Waters DJ, Timms B. Human prostate cancer risk factors. Cancer 2004; 101:2371-490. [PMID: 15495199 DOI: 10.1002/cncr.20408] [Citation(s) in RCA: 398] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prostate cancer has the highest prevalence of any nonskin cancer in the human body, with similar likelihood of neoplastic foci found within the prostates of men around the world regardless of diet, occupation, lifestyle, or other factors. Essentially all men with circulating androgens will develop microscopic prostate cancer if they live long enough. This review is a contemporary and comprehensive, literature-based analysis of the putative risk factors for human prostate cancer, and the results were presented at a multidisciplinary consensus conference held in Crystal City, Virginia, in the fall of 2002. The objectives were to evaluate known environmental factors and mechanisms of prostatic carcinogenesis and to identify existing data gaps and future research needs. The review is divided into four sections, including 1) epidemiology (endogenous factors [family history, hormones, race, aging and oxidative stress] and exogenous factors [diet, environmental agents, occupation and other factors, including lifestyle factors]); 2) animal and cell culture models for prediction of human risk (rodent models, transgenic models, mouse reconstitution models, severe combined immunodeficiency syndrome mouse models, canine models, xenograft models, and cell culture models); 3) biomarkers in prostate cancer, most of which have been tested only as predictive factors for patient outcome after treatment rather than as risk factors; and 4) genotoxic and nongenotoxic mechanisms of carcinogenesis. The authors conclude that most of the data regarding risk relies, of necessity, on epidemiologic studies, but animal and cell culture models offer promise in confirming some important findings. The current understanding of biomarkers of disease and risk factors is limited. An understanding of the risk factors for prostate cancer has practical importance for public health research and policy, genetic and nutritional education and chemoprevention, and prevention strategies.
Collapse
|
50
|
Easton DF, Schaid DJ, Whittemore AS, Isaacs WJ. Where are the prostate cancer genes?--A summary of eight genome wide searches. Prostate 2003; 57:261-9. [PMID: 14601022 DOI: 10.1002/pros.10300] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is strong evidence for genetic susceptibility to prostate cancer, but most of the genes underlying this susceptibility remain to be identified. METHODS We reviewed the results of eight genome-wide linkage searches based on 1,293 families with multiple cases of prostate cancer. RESULTS Across these studies, 11 linkage peaks with LOD scores in excess of 2 were identified. However, no chromosomal region was reported as significant at this level by more than one study and only one corresponded to a peak previously suggested by another group. CONCLUSIONS These results indicate that prostate cancer is genetically complex, and that combined analyses of large family sets will be required to evaluate reliably the linkage evidence.
Collapse
Affiliation(s)
- Douglas F Easton
- Cancer Research U.K. Genetic Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, United Kingdom.
| | | | | | | |
Collapse
|