51
|
Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem 2014; 289:33629-43. [PMID: 25301952 DOI: 10.1074/jbc.m114.589556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.
Collapse
Affiliation(s)
- Sarah E Brennan-Laun
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and
| | - Xiao-Ling Li
- the Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather J Ezelle
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| | | | - Perry J Blackshear
- the Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Gerald M Wilson
- From the Marlene and Stewart Greenebaum Cancer Center, Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Bret A Hassel
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| |
Collapse
|
52
|
Singaraja RR, Tietjen I, Hovingh GK, Franchini PL, Radomski C, Wong K, vanHeek M, Stylianou IM, Lin L, Wang L, Mitnaul L, Hubbard B, Winther M, Mattice M, Legendre A, Sherrington R, Kastelein JJ, Akinsanya K, Plump A, Hayden MR. Identification of four novel genes contributing to familial elevated plasma HDL cholesterol in humans. J Lipid Res 2014; 55:1693-701. [PMID: 24891332 PMCID: PMC4109763 DOI: 10.1194/jlr.m048710] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/01/2014] [Indexed: 01/15/2023] Open
Abstract
While genetic determinants strongly influence HDL cholesterol (HDLc) levels, most genetic causes underlying variation in HDLc remain unknown. We aimed to identify novel rare mutations with large effects in candidate genes contributing to extreme HDLc in humans, utilizing family-based Mendelian genetics. We performed next-generation sequencing of 456 candidate HDLc-regulating genes in 200 unrelated probands with extremely low (≤10th percentile) or high (≥90th percentile) HDLc. Probands were excluded if known mutations existed in the established HDLc-regulating genes ABCA1, APOA1, LCAT, cholesteryl ester transfer protein (CETP), endothelial lipase (LIPG), and UDP-N-acetyl-α-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 2 (GALNT2). We identified 93 novel coding or splice-site variants in 72 candidate genes. Each variant was genotyped in the proband's family. Family-based association analyses were performed for variants with sufficient power to detect significance at P < 0.05 with a total of 627 family members being assessed. Mutations in the genes glucokinase regulatory protein (GCKR), RNase L (RNASEL), leukocyte immunoglobulin-like receptor 3 (LILRA3), and dynein axonemal heavy chain 10 (DNAH10) segregated with elevated HDLc levels in families, while no mutations associated with low HDLc. Taken together, we have identified mutations in four novel genes that may play a role in regulating HDLc levels in humans.
Collapse
Affiliation(s)
- Roshni R. Singaraja
- Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
- A*STAR Institute and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ian Tietjen
- Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - G. Kees Hovingh
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | - Linus Lin
- Merck Research Laboratories, Rahway, NJ
| | | | | | | | | | | | | | | | - John J. Kastelein
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Michael R. Hayden
- A*STAR Institute and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Molecular Medicine and Therapeutics, and Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
53
|
Demichelis F, Stanford JL. Genetic predisposition to prostate cancer: Update and future perspectives. Urol Oncol 2014; 33:75-84. [PMID: 24996773 DOI: 10.1016/j.urolonc.2014.04.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Prostate cancer is the second most frequent cancer in men worldwide and kills over 250,000 men worldwide every year. Prostate cancer is a heterogeneous disease at the clinical and the molecular level. The Scandinavian Twin Registry Study demonstrated that in contrast to most malignancies where environment was the overriding influence, heritable factors account for more than fifty percent of prostate cancers. METHODS AND MATERIALS We review the literature on prostate cancer risk variants (rare and common) including SNPs and Copy Number Variants (CNVs) and discuss the potential implications of significant variants for prostate cancer patient care. RESULTS The search for prostate cancer susceptibility genes has included both family-based studies and case-control studies utilizing a variety of approaches from array-based to sequencing-based studies. A major challenge is to identify genetic variants associated with more aggressive, potentially lethal prostate cancer and to understand their role in the progression of the disease. CONCLUSION Future risk models useful in the clinical setting will likely incorporate several risk loci rather than single variants and may be dependent on an individual patient's ethnic background.
Collapse
Affiliation(s)
- Francesca Demichelis
- Centre for Integrative Biology, University of Trento, Trento, Italy; Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, NY; Institute for Precision Medicine, Weill Medical College of Cornell University and New York Presbyterian Hospital, New York, NY.
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| |
Collapse
|
54
|
Huang H, Zeqiraj E, Dong B, Jha BK, Duffy NM, Orlicky S, Thevakumaran N, Talukdar M, Pillon MC, Ceccarelli DF, Wan LCK, Juang YC, Mao DYL, Gaughan C, Brinton MA, Perelygin AA, Kourinov I, Guarné A, Silverman RH, Sicheri F. Dimeric structure of pseudokinase RNase L bound to 2-5A reveals a basis for interferon-induced antiviral activity. Mol Cell 2014; 53:221-34. [PMID: 24462203 PMCID: PMC3974923 DOI: 10.1016/j.molcel.2013.12.025] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/21/2013] [Accepted: 12/19/2013] [Indexed: 02/01/2023]
Abstract
RNase L is an ankyrin repeat domain-containing dual endoribonuclease-pseudokinase that is activated by unusual 2,′5′-oligoadenylate (2-5A) second messengers and which impedes viral infections in higher vertebrates. Despite its importance in interferon-regulated antiviral innate immunity, relatively little is known about its precise mechanism of action. Here we present a functional characterization of 2.5 Å and 3.25 Å X-ray crystal and small-angle X-ray scattering structures of RNase L bound to a natural 2-5A activator with and without ADP or the nonhydrolysable ATP mimetic AMP-PNP. These studies reveal how recognition of 2-5A through interactions with the ankyrin repeat domain and the pseudokinase domain, together with nucleotide binding, imposes a rigid intertwined dimer configuration that is essential for RNase catalytic and antiviral functions. The involvement of the pseudokinase domain of RNase L in 2-5A sensing, nucleotide binding, dimerization, and ribonuclease functions highlights the evolutionary adaptability of the eukaryotic protein kinase fold. Structural basis for RNase L regulation by 2-5A and nucleotide (ADP or ATP) binding Recognition of 2-5A is mediated by both ankyrin repeat and protein kinase domains Nucleotide enforces a closed conformation of the kinase domain Nucleotide binding to the pseudokinase domain is essential for RNA cleavage function
Collapse
Affiliation(s)
- Hao Huang
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Elton Zeqiraj
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Beihua Dong
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Babal Kant Jha
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nicole M Duffy
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Stephen Orlicky
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Neroshan Thevakumaran
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Manisha Talukdar
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Monica C Pillon
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Derek F Ceccarelli
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Leo C K Wan
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yu-Chi Juang
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Daniel Y L Mao
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Christina Gaughan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Margo A Brinton
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | | - Igor Kourinov
- NE-CAT APS, Building 436E, Argonne National Lab, Argonne, IL 60439, USA
| | - Alba Guarné
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Robert H Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Frank Sicheri
- Program in Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
55
|
|
56
|
Hoeh MP, Deane LA. PSA Screening: A Discussion Based on the USPSTF Recommendations and the AUA and EAU Guidelines. JOURNAL OF MEN'S HEALTH 2014. [DOI: 10.1089/jomh.2014.3502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
57
|
Menegaux F, Anger A, Randrianasolo H, Mulot C, Laurent-Puig P, Iborra F, Bringer JP, Leizour B, Thuret R, Lamy PJ, Rébillard X, Trétarre B. Epidemiological study of prostate cancer (EPICAP): a population-based case-control study in France. BMC Cancer 2014; 14:106. [PMID: 24552491 PMCID: PMC3936778 DOI: 10.1186/1471-2407-14-106] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 02/11/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prostate cancer is the most common cancer in male in most Western countries, including France. Despite a significant morbidity and mortality to a lesser extent, the etiology of prostate cancer remains largely unknown. Indeed, the only well-established risk factors to date are age, ethnicity and a family history of prostate cancer. We present, here, the rationale and design of the EPIdemiological study of Prostate CAncer (EPICAP), a population-based case-control study specifically designed to investigate the role of environmental and genetic factors in prostate cancer. The EPICAP study will particularly focused on the role of circadian disruption, chronic inflammation, hormonal and metabolic factors in the occurrence of prostate cancer. METHODS/DESIGN EPICAP is a population-based case-control study conducted in the département of Hérault in France. Eligible cases are all cases of prostate cancers newly diagnosed in 2012-2013 in men less than 75 years old and residing in the département of Hérault at the time of diagnosis. Controls are men of the same age as the cases and living in the département of Hérault, recruited in the general population.The sample will include a total of 1000 incident cases of prostate cancer and 1000 population-based controls over a 3-year period (2012-2014).The cases and controls are face-to-face interviewed using a standardized computed assisted questionnaire. The questions focus primarily on usual socio-demographic characteristics, personal and family medical history, lifestyle, leisure activities, residential and occupational history. Anthropometric measures and biological samples are also collected for cases and controls. DISCUSSION The EPICAP study aims to answer key questions in prostate cancer etiology: (1) role of circadian disruption through the study of working hours, chronotype and duration/quality of sleep, (2) role of chronic inflammation and anti-inflammatory drugs, (3) role of hormonal and metabolic factors through a detailed questionnaire, (4) role of individual genetic susceptibility of genes involved in biological pathways of interest. The EPICAP study will also allow us to study prognostic factors and tumor aggressiveness.Taken together, the EPICAP study will provide a comprehensive framework to go further in the understanding of prostate cancer occurrence and its prognosis.
Collapse
Affiliation(s)
- Florence Menegaux
- INSERM U1018, Center for Research in Epidemiology and Population Health (CESP), U1018, Environmental Epidemiology of Cancer Team, 16av, Paul Vaillant Couturier, 94807 Villejuif Cédex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Evolutionary conservation and expression of human RNA-binding proteins and their role in human genetic disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:1-55. [PMID: 25201102 DOI: 10.1007/978-1-4939-1221-6_1] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RNA-binding proteins (RBPs) are effectors and regulators of posttranscriptional gene regulation (PTGR). RBPs regulate stability, maturation, and turnover of all RNAs, often binding thousands of targets at many sites. The importance of RBPs is underscored by their dysregulation or mutations causing a variety of developmental and neurological diseases. This chapter globally discusses human RBPs and provides a brief introduction to their identification and RNA targets. We review RBPs based on common structural RNA-binding domains, study their evolutionary conservation and expression, and summarize disease associations of different RBP classes.
Collapse
|
59
|
Nelson WG, Demarzo AM, Yegnasubramanian S. The diet as a cause of human prostate cancer. Cancer Treat Res 2014; 159:51-68. [PMID: 24114474 DOI: 10.1007/978-3-642-38007-5_4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asymptomatic prostate inflammation and prostate cancer have reached epidemic proportions among men in the developed world. Animal model studies implicate dietary carcinogens, such as the heterocyclic amines from over-cooked meats and sex steroid hormones, particularly estrogens, as candidate etiologies for prostate cancer. Each acts by causing epithelial cell damage, triggering an inflammatory response that can evolve into a chronic or recurrent condition. This milieu appears to spawn proliferative inflammatory atrophy (PIA) lesions, a type of focal atrophy that represents the earliest of prostate cancer precursor lesions. Rare PIA lesions contain cells which exhibit high c-Myc expression, shortened telomere segments, and epigenetic silencing of genes such as GSTP1, encoding the π-class glutathione S-transferase, all characteristic of prostatic intraepithelial neoplasia (PIN) and prostate cancer. Subsequent genetic changes, such as the gene translocations/deletions that generate fusion transcripts between androgen-regulated genes (such as TMPRSS2) and genes encoding ETS family transcription factors (such as ERG1), arise in PIN lesions and may promote invasiveness characteristic of prostatic adenocarcinoma cells. Lethal prostate cancers contain markedly corrupted genomes and epigenomes. Epigenetic silencing, which seems to arise in response to the inflamed microenvironment generated by dietary carcinogens and/or estrogens as part of an epigenetic "catastrophe" affecting hundreds of genes, persists to drive clonal evolution through metastatic dissemination. The cause of the initial epigenetic "catastrophe" has not been determined but likely involves defective chromatin structure maintenance by over-exuberant DNA methylation or histone modification. With dietary carcinogens and estrogens driving pro-carcinogenic inflammation in the developed world, it is tempting to speculate that dietary components associated with decreased prostate cancer risk, such as intake of fruits and vegetables, especially tomatoes and crucifers, might act to attenuate the ravages of the chronic or recurrent inflammatory processes. Specifically, nutritional agents might prevent PIA lesions or reduce the propensity of PIA lesions to suffer "catastrophic" epigenome corruption.
Collapse
Affiliation(s)
- William G Nelson
- Departments of Oncology, Pathology, and Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Weinberg Bldg 1100, 1650 Orleans Street, Baltimore, MD, 21231, USA,
| | | | | |
Collapse
|
60
|
Zhang A, Dong B, Doucet AJ, Moldovan JB, Moran JV, Silverman RH. RNase L restricts the mobility of engineered retrotransposons in cultured human cells. Nucleic Acids Res 2013; 42:3803-20. [PMID: 24371271 PMCID: PMC3973342 DOI: 10.1093/nar/gkt1308] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Retrotransposons are mobile genetic elements, and their mobility can lead to genomic instability. Retrotransposon insertions are associated with a diverse range of sporadic diseases, including cancer. Thus, it is not a surprise that multiple host defense mechanisms suppress retrotransposition. The 2′,5′-oligoadenylate (2-5A) synthetase (OAS)-RNase L system is a mechanism for restricting viral infections during the interferon antiviral response. Here, we investigated a potential role for the OAS-RNase L system in the restriction of retrotransposons. Expression of wild type (WT) and a constitutively active form of RNase L (NΔ385), but not a catalytically inactive RNase L mutant (R667A), impaired the mobility of engineered human LINE-1 (L1) and mouse intracisternal A-type particle retrotransposons in cultured human cells. Furthermore, WT RNase L, but not an inactive RNase L mutant (R667A), reduced L1 RNA levels and subsequent expression of the L1-encoded proteins (ORF1p and ORF2p). Consistently, confocal immunofluorescent microscopy demonstrated that WT RNase L, but not RNase L R667A, prevented formation of L1 cytoplasmic foci. Finally, siRNA-mediated depletion of endogenous RNase L in a human ovarian cancer cell line (Hey1b) increased the levels of L1 retrotransposition by ∼2-fold. Together, these data suggest that RNase L might function as a suppressor of structurally distinct retrotransposons.
Collapse
Affiliation(s)
- Ao Zhang
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, 44195, USA, Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA, Department of Human Genetics, Ann Arbor, MI 48109, USA, Cellular and Molecular Biology Program, Ann Arbor, MI 48109, USA, Department of Internal Medicine, Ann Arbor, MI 48109, USA and Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, Michigan, 48109, USA
| | | | | | | | | | | |
Collapse
|
61
|
Genetic analysis of the principal genes related to prostate cancer: A review. Urol Oncol 2013; 31:1419-29. [DOI: 10.1016/j.urolonc.2012.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/27/2012] [Accepted: 07/20/2012] [Indexed: 12/20/2022]
|
62
|
Sorgeloos F, Jha BK, Silverman RH, Michiels T. Evasion of antiviral innate immunity by Theiler's virus L* protein through direct inhibition of RNase L. PLoS Pathog 2013; 9:e1003474. [PMID: 23825954 PMCID: PMC3694852 DOI: 10.1371/journal.ppat.1003474] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/17/2013] [Indexed: 01/08/2023] Open
Abstract
Theiler's virus is a neurotropic picornavirus responsible for chronic infections of the central nervous system. The establishment of a persistent infection and the subsequent demyelinating disease triggered by the virus depend on the expression of L*, a viral accessory protein encoded by an alternative open reading frame of the virus. We discovered that L* potently inhibits the interferon-inducible OAS/RNase L pathway. The antagonism of RNase L by L* was particularly prominent in macrophages where baseline oligoadenylate synthetase (OAS) and RNase L expression levels are elevated, but was detectable in fibroblasts after IFN pretreatment. L* mutations significantly affected Theiler's virus replication in primary macrophages derived from wild-type but not from RNase L-deficient mice. L* counteracted the OAS/RNase L pathway through direct interaction with the ankyrin domain of RNase L, resulting in the inhibition of this enzyme. Interestingly, RNase L inhibition was species-specific as Theiler's virus L* protein blocked murine RNase L but not human RNase L or RNase L of other mammals or birds. Direct RNase L inhibition by L* and species specificity were confirmed in an in vitro assay performed with purified proteins. These results demonstrate a novel viral mechanism to elude the antiviral OAS/RNase L pathway. By targeting the effector enzyme of this antiviral pathway, L* potently inhibits RNase L, underscoring the importance of this enzyme in innate immunity against Theiler's virus. Theiler's virus is a murine picornavirus (same family as poliovirus) which has a striking ability to establish persistent infections of the central nervous system. To do so, the virus has to counteract the immune response of the host and particularly the potent response mediated by interferon. We observed that a protein encoded by Theiler's virus, the L* protein, inhibited the RNase L pathway, one of the best-characterized pathways mediating the antiviral IFN response. In contrast to previously identified viral antagonists of this pathway, L* was found to act directly on RNase L, the effector enzyme of the pathway. L* activity was found to be species-specific as it inhibited murine but not human RNase L. We confirmed the species-specificity and the direct interaction between L* and RNase L in vitro, using purified proteins. Acting at the effector step in the pathway allows L* to block RNase L activity efficiently. This suggests that RNase L is particularly important to control Theiler's virus replication in vivo. Another virus, mouse hepatitis virus (MHV), was recently shown to interfere with RNase L activation. Theiler's virus and MHV share a marked tropism for macrophages which may suggest that the RNase L pathway is particularly important in this cell type.
Collapse
Affiliation(s)
| | - Babal Kant Jha
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio United States of America
| | - Robert H. Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio United States of America
| | - Thomas Michiels
- Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
- * E-mail:
| |
Collapse
|
63
|
Mazaris E, Tsiotras A. Molecular pathways in prostate cancer. Nephrourol Mon 2013; 5:792-800. [PMID: 24282788 PMCID: PMC3830904 DOI: 10.5812/numonthly.9430] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 12/17/2012] [Indexed: 01/02/2023] Open
Abstract
Objectives Prostate cancer is a prevalent disease with a high impact on patients’ morbidity and mortality. Despite efforts to profile prostate cancer, the genetic alterations and biological processes that correlate with disease progression remain partially elusive. The purpose of this study is to review the recent evidence relating to the initiation and progression of prostate cancer in relation to the familial correlation of the disease, the genetic aberrations resulting in prostate cancer and the new molecular biology data regarding prostate cancer. Materials and Methods A Medline database search identified all the existing publications on the molecular events associated with the pathogenesis and evolution of prostate cancer. Particular emphasis was given on the specific genetic phenomena associated with prostate cancer. Results Like other cancers, prostate cancer is caused by an accumulation of genetic alterations in a cell that drives it to malignant growth. Specific genes and gene alterations have been suggested to play a role in its development and progression. Aneuploidy, loss of heterozygosity, gene mutations, hypermethylation and inactivation of specific tumour suppressor genes such as GSTpi, APC, MDR1, GPX3 and others have been detected in prostate cancers, but generally only at a low or moderate frequency. The androgen receptor (AR) signalling pathway may play a crucial role in the early development of prostate cancer, as well as in the development of androgen-independent disease that fails to respond to hormone deprivation therapies. Other alterations linked to the transition to hormone-independence include amplification of MYC and increased expression of ERBB2 and BCL2. Inflammatory changes may also contribute to the development of prostate cancer. Conclusion The identification of specific molecular markers for prostate cancer may lead to its earliest detection and better prediction of its behavior. The better understanding of the molecular events affecting prostate cancer progression may result in the introduction of new drugs to target these events thus providing a potential cure and a tool for prevention of this very common disease.
Collapse
Affiliation(s)
| | - Alexios Tsiotras
- Urology Department, Lister Hospital, Stevenage, United Kingdom
- Corresponding author: Alexios Tsiotras, Urology Department, Lister Hospital, Stevenage, United Kingdom. Tel: +44-7580348549, Fax: +44-1438515601, E-mail:
| |
Collapse
|
64
|
Jha BK, Dong B, Nguyen CT, Polyakova I, Silverman RH. Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol Ther 2013; 21:1749-57. [PMID: 23732991 DOI: 10.1038/mt.2013.112] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/26/2013] [Indexed: 12/23/2022] Open
Abstract
The use of lytic viruses to preferentially infect and eliminate cancer cells while sparing normal cells is a promising experimental therapeutic approach for treating cancer. However, the efficacy of oncolytic virotherapy is often limited by two innate immunity pathways, the protein kinase PKR and the 2'-5'-oligoadenylate (OAS)/RNase L systems, which are widely present in many but not all tumor cell types. Previously, we reported that the anticancer drug, sunitinib, an inhibitor of VEGF-R and PDGF-R, has off-target effects against both PKR and RNase L. Here we show that combining sunitinib treatments with infection by an oncolytic virus, vesicular stomatitis virus (VSV), led to the elimination of prostate, breast, and kidney malignant tumors in mice. In contrast, either virus or sunitinib alone slowed tumor progression but did not eliminate tumors. In prostate tumors excised from treated mice, sunitinib decreased levels of the phosphorylated form of translation initiation factor, eIF2-α, a substrate of PKR, by 10-fold while increasing median viral titers by 23-fold. The sunitinib/VSV regimen caused complete and sustained tumor regression in both immunodeficient and immunocompetent animals. Results indicate that transient inhibition of innate immunity with sunitinib enhances oncolytic virotherapy allowing the recovery of tumor-bearing animals.
Collapse
Affiliation(s)
- Babal K Jha
- Department of Cancer Biology, Lerner Research Institute, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
65
|
Xu J, Sun J, Zheng SL. Prostate cancer risk-associated genetic markers and their potential clinical utility. Asian J Androl 2013; 15:314-22. [PMID: 23564047 PMCID: PMC3739659 DOI: 10.1038/aja.2013.42] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 03/16/2013] [Accepted: 03/18/2013] [Indexed: 02/02/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers among men in Western developed countries and its incidence has increased considerably in many other parts of the world, including China. The etiology of PCa is largely unknown but is thought to be multifactorial, where inherited genetics plays an important role. In this article, we first briefly review results from studies of familial aggregation and genetic susceptibility to PCa. We then recap key findings of rare and high-penetrance PCa susceptibility genes from linkage studies in PCa families. We devote a significant portion of this article to summarizing discoveries of common and low-penetrance PCa risk-associated single-nucleotide polymorphisms (SNPs) from genetic association studies in PCa cases and controls, especially those from genome-wide association studies (GWASs). A strong focus of this article is to review the literature on the potential clinical utility of these implicated genetic markers. Most of these published studies described PCa risk estimation using a genetic score derived from multiple risk-associated SNPs and its utility in determining the need for prostate biopsy. Finally, we comment on the newly proposed concept of genetic score; the notion is to treat it as a marker for genetic predisposition, similar to family history, rather than a diagnostic marker to discriminate PCa patients from non-cancer patients. Available evidence to date suggests that genetic score is an objective and better measurement of inherited risk of PCa than family history. Another unique feature of this article is the inclusion of genetic association studies of PCa in Chinese and Japanese populations.
Collapse
Affiliation(s)
- Jianfeng Xu
- Fudan Institute of Urology, Huashan Hospital, Fudan UniversityFudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | | | | |
Collapse
|
66
|
Ledet EM, Hu X, Sartor O, Rayford W, Li M, Mandal D. Characterization of germline copy number variation in high-risk African American families with prostate cancer. Prostate 2013; 73:614-23. [PMID: 23060098 DOI: 10.1002/pros.22602] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 09/10/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND Prostate cancer is a complex multi-allelic disease and the most common malignancy in men. The incidence of prostate cancer in African American men is more than twice as high as that of any other race. Despite the high prevalence of prostate cancer amongst African American men, this population has been under represented in genetic studies of prostate cancer. Although genomic copy number variations (CNVs) have been detected in prostate tumors, this is the first study describing germline CNVs in African American hereditary prostate cancer families. METHODS Ten high-risk African American families with three or more affected individuals and with an early age of onset were recruited. From these families, 37 individuals, including 23 affected males, and 14 unaffected males, were selected for CNV analysis. Array comparative genomic hybridization was used to characterize germline CNVs unique to African American men with hereditary prostate cancer. RESULTS Through common aberration analysis in affected family members; novel CNVs were identified at chromosomes 1p36.13 and 16q23.3. Differential analysis comparing affected and unaffected family members identified 9.4 kb duplication on chromosome 14q32.33 which segregate with prostate cancer patients in these high-risk families. CONCLUSIONS The duplication at 14q32.33 encompasses IGHG3 gene which has been shown to have both significant gains in copy number as well as overexpression in prostate tumors in African Americans. These CNVs may represent a component of genetic predisposition which contributes to the high prevalence and mortality of prostate cancer in African American men.
Collapse
Affiliation(s)
- Elisa M Ledet
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
67
|
Zeegers MP, Nekeman D, Khan HS, van Dijk BAC, Goldbohm RA, Schalken J, Shajahan S, Pearlman A, Oddoux C, van den Brandt PA, Schouten LJ, Ostrer H. Prostate cancer susceptibility genes on 8p21–23 in a Dutch population. Prostate Cancer Prostatic Dis 2013; 16:248-53. [DOI: 10.1038/pcan.2013.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/01/2013] [Accepted: 02/21/2013] [Indexed: 01/03/2023]
|
68
|
Davoodi H, Esmaeili S, Mortazavian A. Effects of Milk and Milk Products Consumption on Cancer: A Review. Compr Rev Food Sci Food Saf 2013. [DOI: 10.1111/1541-4337.12011] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- H. Davoodi
- Dept. of Clinical Nutrition and Dietology; National Nutrition and Food Technology Research Inst.; Faculty of Nutrition Sciences; Food Science and Technology; Shahid Beheshti Univ. of Medical Sciences; P.O. Box 19395-4741; Tehran; Iran
| | - S. Esmaeili
- Young Researchers Club; Varamin-Pishva Branch; Islamic Azad Univ.; Varamin; Iran
| | - A.M. Mortazavian
- Dept. of Food Science and Technology; National Nutrition and Food Technology Research Inst.; Faculty of Nutrition Sciences; Food Science and Technology; Shahid Beheshti Univ. of Medical Sciences; P.O. Box 19395-4741 Tehran; Iran
| |
Collapse
|
69
|
Kluźniak W, Wokołorczyk D, Kashyap A, Jakubowska A, Gronwald J, Huzarski T, Byrski T, Dębniak T, Gołąb A, Gliniewicz B, Sikorski A, Switała J, Borkowski T, Borkowski A, Antczak A, Wojnar L, Przybyła J, Sosnowski M, Małkiewicz B, Zdrojowy R, Sikorska-Radek P, Matych J, Wilkosz J, Różański W, Kiś J, Bar K, Bryniarski P, Paradysz A, Jersak K, Niemirowicz J, Słupski P, Jarzemski P, Skrzypczyk M, Dobruch J, Domagała P, Akbari MR, Lubiński J, Narod SA, Cybulski C. The G84E mutation in the HOXB13 gene is associated with an increased risk of prostate cancer in Poland. Prostate 2013; 73:542-8. [PMID: 23334858 DOI: 10.1002/pros.22594] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/04/2012] [Indexed: 01/19/2023]
Abstract
BACKGROUND The G84E mutation in the HOXB13 gene has been associated with a high lifetime risk of prostate cancer in North America (about 20-fold). The geographical and ethnic extent of this recurrent allele has not yet been determined. METHODS We assayed for the presence of the G84E mutation in 3,515 prostate cancer patients and 2,604 controls from Poland and estimated the odds ratio for prostate cancer associated with the allele. RESULTS The G84E mutation was detected in 3 of 2,604 (0.1%) individuals from the general population in Poland and in 20 of 3,515 (0.6%) men with prostate cancer (Odds ratio [OR] = 5.0; 95% CI: 1.5-16.7; P = 0.008). The allele was present in 4 of 416 (1.0%) men with familial prostate cancer (OR = 8.4, 95% CI: 1.9-37.7; P = 0.005). CONCLUSIONS The G84E mutation predisposes to prostate cancer in Poland, but accounts for only a small proportion of cases. We expect that the G84E founder mutation might be present in other Slavic populations.
Collapse
Affiliation(s)
- Wojciech Kluźniak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Schoenfeld JD, Margalit DN, Kasperzyk JL, Shui IM, Rider JR, Epstein MM, Meisner A, Kenfield SA, Martin NE, Nguyen PL, Kantoff PW, Giovannucci EL, Stampfer MJ, Mucci LA. A single nucleotide polymorphism in inflammatory gene RNASEL predicts outcome after radiation therapy for localized prostate cancer. Clin Cancer Res 2013; 19:1612-9. [PMID: 23382116 PMCID: PMC3602407 DOI: 10.1158/1078-0432.ccr-12-2718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE To study associations between single nucleotide polymorphisms (SNP) in Ribonuclease L (RNASEL), a gene implicated in inflammation and prostate cancer risk, and outcomes after radiation therapy. EXPERIMENTAL DESIGN We followed participants in the prospective US Health Professionals Follow-Up Study treated with radiation therapy for early-stage prostate cancer. Three SNPs were genotyped based on previously determined functional and biological significance. We used multivariable Cox proportional hazards models to assess per-allele associations with the primary outcome defined as time to a composite endpoint including development of lethal prostate cancer or biochemical recurrence. RESULTS We followed 434 patients treated with radiation therapy for a median of 9 years. On multivariate analysis, the rs12757998 variant allele was associated with significantly decreased risk of the composite endpoint [HR: 0.65; 95% confidence interval (CI), 0.45-0.94%; P = 0.02] driven by decreased biochemical recurrence (HR: 0.60; 95% CI, 0.40-0.89%; P = 0.01) and men treated with external beam (HR: 0.58; 95% CI, 0.36-0.93%; P = 0.02). In contrast, in 516 men from the same cohort treated with radical prostatectomy, we found no significant impact of this variant on outcome. Furthermore, the rs12757998 variant allele significantly modified the association between androgen deprivation therapy and outcomes after radiation therapy (p-interaction = 0.02). CONCLUSION We show an association between RNASEL SNP rs12757998 and outcome after radiation therapy for prostate cancer. This SNP is associated with increased circulating C-reactive protein and interleukin-6, suggesting a potential role for inflammation in the response to radiation. If validated, genetic predictors of outcome may help inform prostate cancer management.
Collapse
|
71
|
de Mesquita Netto AC, Gomez RS, Diniz MG, Fonseca-Silva T, Campos K, De Marco L, Carlos R, Gomes CC. Assessing the contribution of HRPT2 to the pathogenesis of jaw fibrous dysplasia, ossifying fibroma, and osteosarcoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2013; 115:359-67. [DOI: 10.1016/j.oooo.2012.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 11/09/2012] [Accepted: 11/16/2012] [Indexed: 10/27/2022]
|
72
|
Laitinen VH, Wahlfors T, Saaristo L, Rantapero T, Pelttari LM, Kilpivaara O, Laasanen SL, Kallioniemi A, Nevanlinna H, Aaltonen L, Vessella RL, Auvinen A, Visakorpi T, Tammela TLJ, Schleutker J. HOXB13 G84E mutation in Finland: population-based analysis of prostate, breast, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 2013; 22:452-60. [PMID: 23292082 DOI: 10.1158/1055-9965.epi-12-1000-t] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND A recently identified germline mutation G84E in HOXB13 was shown to increase the risk of prostate cancer. In a family-based analysis by The International Consortium for Prostate Cancer Genetics (ICPCG), the G84E mutation was most prevalent in families from the Nordic countries of Finland (22.4%) and Sweden (8.2%). METHODS To further investigate the importance of G84E in the Finns, we determined its frequency in more than 4,000 prostate cancer cases and 5,000 controls. In addition, 986 breast cancer and 442 colorectal cancer (CRC) cases were studied. Genotyping was conducted using TaqMan, MassARRAY iPLEX, and sequencing. Statistical analyses were conducted using Fisher exact test, and overall survival was analyzed using Cox modeling. RESULTS The frequency of the G84E mutation was significantly higher among patients with prostate cancer and highest among patients with a family history of the disease, hereditary prostate cancer [8.4% vs. 1.0% in controls; OR 8.8; 95% confidence interval (CI), 4.9-15.7]. The mutation contributed significantly to younger age (≤55 years) at onset and high prostate-specific antigen (PSA; ≥20 ng/mL) at diagnosis. An association with increased prostate cancer risk in patients with prior benign prostate hyperplasia (BPH) diagnosis was also revealed. No statistically significant evidence for a contribution in CRC risk was detected, but a suggestive role for the mutation was observed in familial BRCA1/2-negative breast cancer. CONCLUSIONS These findings confirm an increased cancer risk associated with the G84E mutation in the Finnish population, particularly for early-onset prostate cancer and cases with substantially elevated PSA. IMPACT This study confirms the overall importance of the HOXB13 G84E mutation in prostate cancer susceptibility.
Collapse
Affiliation(s)
- Virpi H Laitinen
- Johanna Schleutker, Medical Biochemistry and Genetics, Institute of Biomedicine, Kiinamyllynkatu 10, FI-20014 University of Turku, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
The Role of Cholesterol in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
74
|
Xu J, Lange EM, Lu L, Zheng SL, Wang Z, Thibodeau SN, Cannon-Albright LA, Teerlink CC, Camp NJ, Johnson AM, Zuhlke KA, Stanford JL, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Maier C, Luedeke M, Vogel W, Schleutker J, Wahlfors T, Tammela T, Schaid D, McDonnell SK, DeRycke MS, Cancel-Tassin G, Cussenot O, Wiklund F, Grönberg H, Eeles R, Easton D, Kote-Jarai Z, Whittemore AS, Hsieh CL, Giles GG, Hopper JL, Severi G, Catalona WJ, Mandal D, Ledet E, Foulkes WD, Hamel N, Mahle L, Moller P, Powell I, Bailey-Wilson JE, Carpten JD, Seminara D, Cooney KA, Isaacs WB, International Consortium for Prostate Cancer Genetics. HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG). Hum Genet 2013; 132:5-14. [PMID: 23064873 PMCID: PMC3535370 DOI: 10.1007/s00439-012-1229-4] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/15/2012] [Indexed: 11/26/2022]
Abstract
Prostate cancer has a strong familial component but uncovering the molecular basis for inherited susceptibility for this disease has been challenging. Recently, a rare, recurrent mutation (G84E) in HOXB13 was reported to be associated with prostate cancer risk. Confirmation and characterization of this finding is necessary to potentially translate this information to the clinic. To examine this finding in a large international sample of prostate cancer families, we genotyped this mutation and 14 other SNPs in or flanking HOXB13 in 2,443 prostate cancer families recruited by the International Consortium for Prostate Cancer Genetics (ICPCG). At least one mutation carrier was found in 112 prostate cancer families (4.6 %), all of European descent. Within carrier families, the G84E mutation was more common in men with a diagnosis of prostate cancer (194 of 382, 51 %) than those without (42 of 137, 30 %), P = 9.9 × 10(-8) [odds ratio 4.42 (95 % confidence interval 2.56-7.64)]. A family-based association test found G84E to be significantly over-transmitted from parents to affected offspring (P = 6.5 × 10(-6)). Analysis of markers flanking the G84E mutation indicates that it resides in the same haplotype in 95 % of carriers, consistent with a founder effect. Clinical characteristics of cancers in mutation carriers included features of high-risk disease. These findings demonstrate that the HOXB13 G84E mutation is present in ~5 % of prostate cancer families, predominantly of European descent, and confirm its association with prostate cancer risk. While future studies are needed to more fully define the clinical utility of this observation, this allele and others like it could form the basis for early, targeted screening of men at elevated risk for this common, clinically heterogeneous cancer.
Collapse
Affiliation(s)
- Jianfeng Xu
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Ethan M. Lange
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Departments of Genetics and Biostatistics, University of North Carolina, Chapel Hill, NC USA
| | - Lingyi Lu
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Siqun L. Zheng
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Zhong Wang
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Stephen N. Thibodeau
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Lisa A. Cannon-Albright
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Craig C. Teerlink
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Nicola J. Camp
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Anna M. Johnson
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - Kimberly A. Zuhlke
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - Janet L. Stanford
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Division of Public Health Sciences, FHCRC, Seattle, WA USA
| | - Elaine A. Ostrander
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Kathleen E. Wiley
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Sarah D. Isaacs
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Patrick C. Walsh
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Christiane Maier
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
| | - Manuel Luedeke
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
| | - Walther Vogel
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Johanna Schleutker
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Tiina Wahlfors
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
| | - Teuvo Tammela
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Daniel Schaid
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Shannon K. McDonnell
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Melissa S. DeRycke
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | | | - Olivier Cussenot
- CeRePP ICPCG Group, Paris, France
- Department of Urology, APHP, Hospital Tenon, Paris, France
| | - Fredrik Wiklund
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ros Eeles
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Doug Easton
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Strangeways Laboratory, Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Zsofia Kote-Jarai
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Alice S. Whittemore
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA USA
- Stanford Comprehensive Cancer Center, Stanford School of Medicine, Stanford, CA USA
| | - Chih-Lin Hsieh
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Urology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA USA
| | - Graham G. Giles
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - John L. Hopper
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - Gianluca Severi
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - William J. Catalona
- Northwestern University ICPCG Group, Chicago, IL USA
- Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Diptasri Mandal
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
| | - Elisa Ledet
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
| | - William D. Foulkes
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Nancy Hamel
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Lovise Mahle
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- The Norwegian Radium Hospital, Oslo, Norway
| | - Pal Moller
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- The Norwegian Radium Hospital, Oslo, Norway
| | - Isaac Powell
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
| | - Joan E. Bailey-Wilson
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Inherited Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - John D. Carpten
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Genetic Basis of Human Disease Research Division, Translational Genomics Research Institute, Phoenix, AZ USA
| | | | - Kathleen A. Cooney
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - William B. Isaacs
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - International Consortium for Prostate Cancer Genetics
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Departments of Genetics and Biostatistics, University of North Carolina, Chapel Hill, NC USA
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Division of Public Health Sciences, FHCRC, Seattle, WA USA
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
- CeRePP ICPCG Group, Paris, France
- Department of Urology, APHP, Hospital Tenon, Paris, France
- CeRePP UPMC University, Paris, France
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
- Strangeways Laboratory, Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA USA
- Stanford Comprehensive Cancer Center, Stanford School of Medicine, Stanford, CA USA
- Department of Urology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA USA
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
- Northwestern University ICPCG Group, Chicago, IL USA
- Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
- The Norwegian Radium Hospital, Oslo, Norway
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
- Inherited Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
- Genetic Basis of Human Disease Research Division, Translational Genomics Research Institute, Phoenix, AZ USA
- National Cancer Institute, NIH, Bethesda, MD USA
| |
Collapse
|
75
|
|
76
|
Lee D, Das Gupta J, Gaughan C, Steffen I, Tang N, Luk KC, Qiu X, Urisman A, Fischer N, Molinaro R, Broz M, Schochetman G, Klein EA, Ganem D, DeRisi JL, Simmons G, Hackett J, Silverman RH, Chiu CY. In-depth investigation of archival and prospectively collected samples reveals no evidence for XMRV infection in prostate cancer. PLoS One 2012; 7:e44954. [PMID: 23028701 PMCID: PMC3445615 DOI: 10.1371/journal.pone.0044954] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 08/10/2012] [Indexed: 12/31/2022] Open
Abstract
XMRV, or xenotropic murine leukemia virus (MLV)-related virus, is a novel gammaretrovirus originally identified in studies that analyzed tissue from prostate cancer patients in 2006 and blood from patients with chronic fatigue syndrome (CFS) in 2009. However, a large number of subsequent studies failed to confirm a link between XMRV infection and CFS or prostate cancer. On the contrary, recent evidence indicates that XMRV is a contaminant originating from the recombination of two mouse endogenous retroviruses during passaging of a prostate tumor xenograft (CWR22) in mice, generating laboratory-derived cell lines that are XMRV-infected. To confirm or refute an association between XMRV and prostate cancer, we analyzed prostate cancer tissues and plasma from a prospectively collected cohort of 39 patients as well as archival RNA and prostate tissue from the original 2006 study. Despite comprehensive microarray, PCR, FISH, and serological testing, XMRV was not detected in any of the newly collected samples or in archival tissue, although archival RNA remained XMRV-positive. Notably, archival VP62 prostate tissue, from which the prototype XMRV strain was derived, tested negative for XMRV on re-analysis. Analysis of viral genomic and human mitochondrial sequences revealed that all previously characterized XMRV strains are identical and that the archival RNA had been contaminated by an XMRV-infected laboratory cell line. These findings reveal no association between XMRV and prostate cancer, and underscore the conclusion that XMRV is not a naturally acquired human infection.
Collapse
Affiliation(s)
- Deanna Lee
- Department of Laboratory Medicine, University of San Francisco, San Francisco, California, United States of America
- University of California San Francisco-Abbott Viral Diagnostics and Discovery Center, University of California San Francisco, San Francisco, California, United States of America
| | | | | | - Imke Steffen
- Blood Systems Research Institute, San Francisco, California, United States of America
| | - Ning Tang
- Abbott Laboratories, Abbott Park, Illinois, United States of America
| | - Ka-Cheung Luk
- Abbott Laboratories, Abbott Park, Illinois, United States of America
| | - Xiaoxing Qiu
- Abbott Laboratories, Abbott Park, Illinois, United States of America
| | - Anatoly Urisman
- Department of Laboratory Medicine, University of San Francisco, San Francisco, California, United States of America
| | - Nicole Fischer
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ross Molinaro
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Miranda Broz
- Department of Laboratory Medicine, University of San Francisco, San Francisco, California, United States of America
| | | | - Eric A. Klein
- Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Don Ganem
- Novartis Institutes for Biomedical Research, Emeryville, California, United States of America
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Graham Simmons
- Blood Systems Research Institute, San Francisco, California, United States of America
| | - John Hackett
- Abbott Laboratories, Abbott Park, Illinois, United States of America
| | | | - Charles Y. Chiu
- Department of Laboratory Medicine, University of San Francisco, San Francisco, California, United States of America
- University of California San Francisco-Abbott Viral Diagnostics and Discovery Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, Division of Infectious Diseases, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
77
|
Bambury RM, Gallagher DJ. Prostate cancer: germline prediction for a commonly variable malignancy. BJU Int 2012; 110:E809-18. [PMID: 22974436 DOI: 10.1111/j.1464-410x.2012.11450.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
UNLABELLED What's known on the subject? and What does the study add? Prostate cancer is a heterogeneous disease and biomarkers to predict its incidence and subsequent clinical behaviour are needed to tailor screening, prevention and therapeutic strategies. Rare mutations in genes such as BRCA1, BRCA2 and HOXB13 can affect prostate cancer incidence and/or clinical behaviour. Genome wide association studies (GWAS) have identified more common genetic variations that explain an estimated 20% of familial prostate cancer risk. In this review, we focus on the potential of germline genetic variation to provide biomarkers for prostate cancer screening, prevention and management. We discuss how germline genetics may have a role in treatment selection if reliable pharmacogenetic predictors of efficacy and toxicity can be identified. We have outlined possible mechanisms for including germline investigation in future prostate cancer clinical trials. OBJECTIVES • Prostate cancer is a heterogeneous disease and biomarkers to predict its incidence and subsequent clinical behaviour are needed to tailor screening, prevention and therapeutic strategies. • In this review we focus on the potential of germline genetic variation to provide these biomarkers. METHODS • We review the published literature on germline genetics in prostate cancer and examine the possibility of including germline genetic biomarkers in future prostate cancer clinical trials. RESULTS • Rare mutations in genes such as BRCA1, BRCA2 and HOXB13 can affect prostate cancer incidence and/or clinical behaviour. • Genome-wide association studies (GWAS) have identified more common genetic variations that explain an estimated 20% of familial prostate cancer risk. • Germline genetics may have a role in treatment selection, if reliable pharmacogenetic predictors of efficacy and toxicity can be identified. CONCLUSION • This rapidly emerging area of prostate cancer research may provide answers to current clinical conundrums in the prostate cancer treatment paradigm. We have outlined possible mechanisms for including germline investigation in future prostate cancer clinical trial design.
Collapse
Affiliation(s)
- Richard M Bambury
- Department of Medical Oncology, Mater Misericordiae University Hospital and St James's Hospital, Dublin, Ireland.
| | | |
Collapse
|
78
|
Genetic heterogeneity in Finnish hereditary prostate cancer using ordered subset analysis. Eur J Hum Genet 2012; 21:437-43. [PMID: 22948022 DOI: 10.1038/ejhg.2012.185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Prostate cancer (PrCa) is the most common male cancer in developed countries and the second most common cause of cancer death after lung cancer. We recently reported a genome-wide linkage scan in 69 Finnish hereditary PrCa (HPC) families, which replicated the HPC9 locus on 17q21-q22 and identified a locus on 2q37. The aim of this study was to identify and to detect other loci linked to HPC. Here we used ordered subset analysis (OSA), conditioned on nonparametric linkage to these loci to detect other loci linked to HPC in subsets of families, but not the overall sample. We analyzed the families based on their evidence for linkage to chromosome 2, chromosome 17 and a maximum score using the strongest evidence of linkage from either of the two loci. Significant linkage to a 5-cM linkage interval with a peak OSA nonparametric allele-sharing LOD score of 4.876 on Xq26.3-q27 (ΔLOD=3.193, empirical P=0.009) was observed in a subset of 41 families weakly linked to 2q37, overlapping the HPCX1 locus. Two peaks that were novel to the analysis combining linkage evidence from both primary loci were identified; 18q12.1-q12.2 (OSA LOD=2.541, ΔLOD=1.651, P=0.03) and 22q11.1-q11.21 (OSA LOD=2.395, ΔLOD=2.36, P=0.006), which is close to HPC6. Using OSA allows us to find additional loci linked to HPC in subsets of families, and underlines the complex genetic heterogeneity of HPC even in highly aggregated families.
Collapse
|
79
|
The association of elevated 2',5'-oligoadenylate-dependent RNase L with lung cancer correlated with deficient enzymatic activity and decreased capacity of RNase L dimerization. Lung Cancer 2012; 78:30-8. [PMID: 22925698 DOI: 10.1016/j.lungcan.2012.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 07/09/2012] [Accepted: 07/30/2012] [Indexed: 01/22/2023]
Abstract
RNase L mediates critical cellular functions including antiviral, proapoptotic, antiproliferative and tumor suppressive activities. In this study, the expression and function of RNase L in lung cancer cells were examined. Interestingly we have found that the expression of RNase L in lung cancer cells was 3- and 9-fold higher in its mRNA and protein levels, but a significant decrease of its enzymatic activity when compared to that in corresponding normal lung cells. Further investigation revealed that 2-5A-induced dimerization of the RNase L protein, a necessary prerequisite for activation of RNase L, was inhibited, as a result of that RLI, a specific inhibitor of RNase L, was remarkably up-regulated in the cancer cells. Our findings provide new insight into how cancer cells escape normal growth-regulating mechanisms to form a tumor and the information may be useful for the design of novel strategies for treating lung cancer through regulating RNase L activity.
Collapse
|
80
|
Genomic Signatures Associated with the Development, Progression, and Outcome of Prostate Cancer. Mol Diagn Ther 2012; 11:345-54. [DOI: 10.1007/bf03256258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
81
|
Lack of evidence for a role of xenotropic murine leukemia virus-related virus in the pathogenesis of prostate cancer and/or chronic fatigue syndrome. Virus Res 2012; 167:1-7. [DOI: 10.1016/j.virusres.2012.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/07/2012] [Accepted: 04/10/2012] [Indexed: 11/18/2022]
|
82
|
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
|
83
|
Al-Haj L, Blackshear PJ, Khabar KSA. Regulation of p21/CIP1/WAF-1 mediated cell-cycle arrest by RNase L and tristetraprolin, and involvement of AU-rich elements. Nucleic Acids Res 2012; 40:7739-52. [PMID: 22718976 PMCID: PMC3439922 DOI: 10.1093/nar/gks545] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The p21Cip1/WAF1 plays an important role in cell-cycle arrest. Here, we find that RNase L regulates p21-mediated G1 growth arrest in AU-rich elements-dependent manner. We found a significant loss of p21 mRNA expression in RNASEL−/− MEFs and that the overexpression of RNase L in HeLa cells induces p21 mRNA expression. The p21 mRNA half-life significantly changes as a result of RNase L modulation, indicating a post-transcriptional effect. Indeed, we found that RNase L promotes tristetraprolin (TTP/ZFP36) mRNA decay. This activity was not seen with dimerization- and nuclease-deficient RNase L mutants. Deficiency in TTP led to increases in p21 mRNA and protein. With induced ablation of RNase L, TTP mRNA and protein expressions were higher, while p21 expression became reduced. We further establish that TTP, but not C124R TTP mutant, binds to, and accelerates the decay of p21 mRNA. The p21 mRNA half-life was prolonged in TTP−/− MEFs. The TTP regulation of p21 mRNA decay required functional AU-rich elements. Thus, we demonstrate a novel mechanism of regulating G1 growth arrest by an RNase L-TTP-p21 axis.
Collapse
Affiliation(s)
- Latifa Al-Haj
- Program in BioMolecular Research, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | | | | |
Collapse
|
84
|
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
|
85
|
Fukuma Y, Matsui H, Koike H, Sekine Y, Shechter I, Ohtake N, Nakata S, Ito K, Suzuki K. Role of squalene synthase in prostate cancer risk and the biological aggressiveness of human prostate cancer. Prostate Cancer Prostatic Dis 2012; 15:339-45. [PMID: 22546838 DOI: 10.1038/pcan.2012.14] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We previously conducted a genome-wide linkage analysis of Japanese nuclear families affected with prostate cancer and showed that the susceptibility to prostate cancer was closely linked to D8S550 at 8p23. The role of farnesyl diphosphate farnesyltransferase (FDFT1), which is located under the peak marker D8S550 at 8p23, and squalene synthase, the enzyme encoded by FDFT1, in prostate cancer was studied. METHODS The association among common variants of FDFT1 with prostate cancer risk, the promoter activities of FDFT1 with different genotypes and the effects of inhibition of squalene synthase were studied, and the FDFT1 transcript levels of human prostate samples were quantified. RESULTS The A allele of rs2645429 was significantly associated with prostate cancer risk in a Japanese familial prostate cancer population. Rs2645429 was located in the promoter region of FDFT1, and the AA genotype showed significantly increased promoter activity. The knockdown of FDFT1 mRNA expression or squalene synthase inhibition led to a significant decrease in prostate cancer cell proliferation. Additionally, human prostate cancer specimens expressed significantly higher levels of FDFT1 mRNA compared with noncancerous specimens. Finally, aggressive cancers showed higher transcript levels. CONCLUSIONS FDFT1 and its encoded enzyme, squalene synthase, may play an important role in prostate cancer development and its aggressive phenotypes.
Collapse
Affiliation(s)
- Y Fukuma
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Jin W, Wu DD, Zhang X, Irwin DM, Zhang YP. Positive selection on the gene RNASEL: correlation between patterns of evolution and function. Mol Biol Evol 2012; 29:3161-8. [PMID: 22513284 DOI: 10.1093/molbev/mss123] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RNASEL is a 2-5A-dependent endoribonuclease that is a component of the interferon-induced 2-5A system, which plays a crucial role in the antiviral and apoptotic activities of interferons. In humans, many polymorphic sites within the RNASEL gene have been associated with an increased risk of developing prostate cancer. Here, we obtained coding sequences for the RNASEL gene from 11 primates and found evidence that positive selection has operated on the C-terminal endoribonuclease domain and the N-terminal ankyrin repeats domain of the protein, domains that directly interact with virus (i.e., ankyrin repeats are responsible for receiving environmental signals, and the endoribonuclease catalyses the destruction of the pathogenic viral RNA). To extend this finding, we studied variation within this gene in modern human populations by resequencing alleles from 144 individuals representing four separate populations. Interestingly, the frequency of the 541D allele shows a negative association with the incidence rate of prostate cancer in worldwide populations, and haplotypes containing the 541D polymorphisms demonstrate signatures of positive selection. RNASEL variants having the 541D haplotype likely have a greater ability to defend against infections by viruses, thus the loss of this activity may be associated with the development of prostate cancer. We provide evidence that positive selection has operated on the RNASEL gene, and its evolution is correlated with its function in pathogen defense and cancer association.
Collapse
Affiliation(s)
- Wei Jin
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| | | | | | | | | |
Collapse
|
87
|
Nakagawa H, Akamatsu S, Takata R, Takahashi A, Kubo M, Nakamura Y. Prostate cancer genomics, biology, and risk assessment through genome-wide association studies. Cancer Sci 2012; 103:607-13. [PMID: 22181854 DOI: 10.1111/j.1349-7006.2011.02193.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 11/22/2011] [Accepted: 12/11/2011] [Indexed: 01/12/2023] Open
Abstract
Prostate cancer (PC) is the most common malignancy observed in men. It is evident that genetic factors play some important roles in PC etiology. Recently, genome-wide association studies in diverse ethnic groups have identified more than 40 germline variants of various genes or chromosomal loci that are significantly associated with PC susceptibility, including multiple 8q24 loci, prostate-specific genes, metabolic and hormone-related genes, and many regions where no coding gene is annotated. However, there are only a few variants or genes for which biological significance or functions have been elucidated so far. The greatest challenge related to genome-wide association studies loci in prostate genomics is to understand the functional consequences of these PC-associated loci and their involvement in PC biology and carcinogenesis. There have been attempts to determine PC risk estimations by combining multiple PC-associated variants for clinical tests, and these can identify a very minor population with high risk of PC. However, they cannot distinguish risk of aggressive PC from that of non-aggressive PC. Further identification of PC-susceptibility loci in larger genome-wide association studies cohorts and biological insights gained from such functional analyses have the potential to translate into clinical benefits, including the development of reliable biomarkers, risk estimation, and effective strategies for screening and prevention of PC.
Collapse
Affiliation(s)
- Hidewaki Nakagawa
- Laboratory for Biomarker Development, Center for Genomic Medicine, RIKEN, Yokohama, Japan.
| | | | | | | | | | | |
Collapse
|
88
|
Sfanos KS, Aloia AL, De Marzo AM, Rein A. XMRV and prostate cancer--a 'final' perspective. Nat Rev Urol 2012; 9:111-8. [PMID: 22231291 DOI: 10.1038/nrurol.2011.225] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
XMRV was first described in 2006, when it was identified in samples isolated from prostate cancer tissues. However, studies have since shown that XMRV arose in the laboratory and was formed by genetic recombination between two viral genomes carried in the germline DNA of mice used during serial transplantation of the CWR22 prostate cancer xenograft. These new findings strongly imply that XMRV does not circulate in humans, but is only present in the laboratory. Thus, there is no reason to believe that it has any role in the etiology of prostate cancer or other diseases.
Collapse
Affiliation(s)
- Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | | | | | | |
Collapse
|
89
|
Ezelle HJ, Hassel BA. Pathologic effects of RNase-L dysregulation in immunity and proliferative control. Front Biosci (Schol Ed) 2012; 4:767-86. [PMID: 22202089 DOI: 10.2741/s298] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The endoribonuclease RNase-L is the terminal component of an RNA cleavage pathway that mediates antiviral, antiproliferative and immunomodulatory activities. Inactivation or dysregulation of RNase-L is associated with a compromised immune response and increased risk of cancer, accordingly its activity is tightly controlled and requires an allosteric activator, 2',5'-linked oligoadenylates, for enzymatic activity. The biological activities of RNase-L are a result of direct and indirect effects of RNA cleavage and microarray analyses have revealed that RNase-L impacts the gene expression program at multiple levels. The identification of RNase-L-regulated RNAs has provided insights into potential mechanisms by which it exerts antiproliferative, proapoptotic, senescence-inducing and innate immune activities. RNase-L protein interactors have been identified that serve regulatory functions and are implicated as alternate mechanisms of its biologic functions. Thus, while the molecular details are understood for only a subset of RNase-L activities, its regulation by small molecules and critical roles in host defense and as a candidate tumor suppressor make it a promising therapeutic target.
Collapse
Affiliation(s)
- Heather J Ezelle
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
90
|
Mandal S, Abebe F, Chaudhary J. 2'-5' oligoadenylate synthetase 1 polymorphism is associated with prostate cancer. Cancer 2011; 117:5509-18. [PMID: 21638280 PMCID: PMC3167978 DOI: 10.1002/cncr.26219] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/25/2011] [Accepted: 04/04/2011] [Indexed: 11/09/2022]
Abstract
BACKGROUND The antiviral, proapoptotic, antiproliferative gene 2'-5' oligoadenylate synthetase (2-5OAS1) converts adenosine triphosphate into a series of 2'-5' oligoadenylates (2-5A). In turn, 2-5A activates latent ribonuclease (RNaseL), a candidate hereditary prostate cancer gene. OAS1 polymorphism (reference single nucleotide polymorphism [SNP] 2660 [rs2660]) has been associated with increased susceptibility to infections and various diseases. In general, the low-enzyme-activity adenine-adenine (AA) genotype promotes susceptibility, whereas the high-enzyme-activity guanosine-guanosine (GG) genotype confers protection. In this study, the authors investigated the association of this functional OAS1 polymorphism (rs2660) with prostate cancer. METHODS Sample size and power were calculated using a power calculation software program for case-control genetic association analyses. Genomic DNA samples from a control group (n = 140) and from a case group of patients with prostate cancer (n = 164) were used for genotyping SNPs rs2660, rs1131454, and rs34137742 in all samples. Statistical analyses were performed using a logistic regression model. RESULTS A significant association was observed between the rs2660 genotype (A/G) and prostate cancer. Genotype AA increased the risk, whereas genotype GG decreased the risk of prostate cancer. The GG genotype was not observed in the African American samples. The AA genotype also increased the risk of prostate cancer with age. CONCLUSIONS The OAS1 SNP rs2660 AA genotype was associated significantly with prostate cancer, whereas the GG genotype protected against prostate cancer. OAS1 rs2660 may be a prostate cancer susceptibility polymorphism, which is a significant observation, especially in a context of the OAS1-RNaseL pathway. Thus, a functional defect in OAS1 because of the rs2660 SNP not only can attenuate RNaseL function but also can alter cell growth and apoptosis independent of RNaseL.
Collapse
Affiliation(s)
- Sanjay Mandal
- Center For Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, Georgia
| | - Fisseha Abebe
- Department of Mathematics, Clark Atlanta University, Atlanta, Georgia
| | - Jaideep Chaudhary
- Center For Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, Georgia
| |
Collapse
|
91
|
|
92
|
Kearney MF, Lee K, Bagni RK, Wiegand A, Spindler J, Maldarelli F, Pinto PA, Linehan WM, Vocke CD, Delviks-Frankenberry KA, deVere White RW, Del Prete GQ, Mellors JW, Lifson JD, KewalRamani VN, Pathak VK, Coffin JM, Le Grice SFJ. Nucleic Acid, Antibody, and Virus Culture Methods to Detect Xenotropic MLV-Related Virus in Human Blood Samples. Adv Virol 2011; 2011:272193. [PMID: 22312339 PMCID: PMC3265299 DOI: 10.1155/2011/272193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/08/2011] [Accepted: 08/27/2011] [Indexed: 11/17/2022] Open
Abstract
The MLV-related retrovirus, XMRV, was recently identified and reported to be associated with both prostate cancer and chronic fatigue syndrome. At the National Cancer Institute-Frederick, MD (NCI-Frederick), we developed highly sensitive methods to detect XMRV nucleic acids, antibodies, and replication competent virus. Analysis of XMRV-spiked samples and/or specimens from two pigtail macaques experimentally inoculated with 22Rv1 cell-derived XMRV confirmed the ability of the assays used to detect XMRV RNA and DNA, and culture isolatable virus when present, along with XMRV reactive antibody responses. Using these assays, we did not detect evidence of XMRV in blood samples (N = 134) or prostate specimens (N = 19) from two independent cohorts of patients with prostate cancer. Previous studies detected XMRV in prostate tissues. In the present study, we primarily investigated the levels of XMRV in blood plasma samples collected from patients with prostate cancer. These results demonstrate that while XMRV-related assays developed at the NCI-Frederick can readily measure XMRV nucleic acids, antibodies, and replication competent virus, no evidence of XMRV was found in the blood of patients with prostate cancer.
Collapse
Affiliation(s)
- M. F. Kearney
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - K. Lee
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - R. K. Bagni
- Protein Expression Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD 21702, USA
| | - A. Wiegand
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - J. Spindler
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - F. Maldarelli
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - P. A. Pinto
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - W. M. Linehan
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - C. D. Vocke
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | - G. Q. Del Prete
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, MD 21702, USA
| | - J. W. Mellors
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - J. D. Lifson
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, MD 21702, USA
| | - V. N. KewalRamani
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - V. K. Pathak
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | - J. M. Coffin
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02155, USA
| | - S. F. J. Le Grice
- HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| |
Collapse
|
93
|
Fesinmeyer MD, Kwon EM, Fu R, Ostrander EA, Stanford JL. Genetic variation in RNASEL and risk for prostate cancer in a population-based case-control study. Prostate 2011; 71:1538-47. [PMID: 21360564 PMCID: PMC3130811 DOI: 10.1002/pros.21370] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 01/31/2011] [Indexed: 01/02/2023]
Abstract
BACKGROUND Linkage studies have implicated chromosome 1q24 as a putative locus for hereditary prostate cancer. The RNASEL gene maps to 1q24 and has been associated with prostate cancer risk in multiple family-based linkage studies. The RNASEL gene product combats viral infection by degrading viral RNA and inducing apoptosis of infected cells. Few studies have evaluated the role of RNASEL variants in unselected or sporadic prostate cancer, or have considered the potential interaction between RNASEL variants and patient characteristics associated with past infection. METHODS Ten SNPs in the RNASEL gene were genotyped in 1,308 prostate cancer cases and 1,267 age-matched controls from prior population-based, case-control studies. The association between each SNP and haplotype with prostate cancer risk was calculated using logistic regression. Associations stratified by Gleason score were evaluated using polytomous regression. The likelihood ratio test was used to investigate effect modification. RESULTS Two RNASEL SNPs were associated with overall increases in prostate cancer risk (OR = 1.13 for each variant allele of rs12723593; OR = 1.88 for any variant allele of rs56250729). Risk estimates did not vary substantially by Gleason score, but there was effect modification for the variant allele of rs635261 by history of prostatitis (P = 0.02). CONCLUSIONS This study identified three RNASEL variants that are associated with risk for prostate cancer. Further research is required to confirm these results and to better understand the potential role RNASEL variants may play in the etiology of sporadic prostate cancer.
Collapse
Affiliation(s)
- Megan D. Fesinmeyer
- Epidemiology Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Erika M. Kwon
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rong Fu
- Epidemiology Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Elaine A. Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Janet L. Stanford
- Epidemiology Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
94
|
Freeman MR, Solomon KR. Cholesterol and benign prostate disease. Differentiation 2011; 82:244-52. [PMID: 21862201 DOI: 10.1016/j.diff.2011.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 04/13/2011] [Accepted: 04/21/2011] [Indexed: 12/17/2022]
Abstract
The origins of benign prostatic diseases, such as benign prostatic hyperplasia (BPH) and chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS), are poorly understood. Patients suffering from benign prostatic symptoms report a substantially reduced quality of life, and the relationship between benign prostate conditions and prostate cancer is uncertain. Epidemiologic data for BPH and CP/CPPS are limited, however an apparent association between BPH symptoms and cardiovascular disease (CVD) has been consistently reported. The prostate synthesizes and stores large amounts of cholesterol and prostate tissues may be particularly sensitive to perturbations in cholesterol metabolism. Hypercholesterolemia, a major risk factor for CVD, is also a risk factor for BPH. Animal model and clinical trial findings suggest that agents that inhibit cholesterol absorption from the intestine, such as the class of compounds known as polyene macrolides, can reduce prostate gland size and improve lower urinary tract symptoms (LUTS). Observational studies indicate that cholesterol-lowering drugs reduce the risk of aggressive prostate cancer, while prostate cancer cell growth and survival pathways depend in part on cholesterol-sensitive biochemical mechanisms. Here we review the evidence that cholesterol metabolism plays a role in the incidence of benign prostate disease and we highlight possible therapeutic approaches based on this concept.
Collapse
Affiliation(s)
- Michael R Freeman
- The Urological Diseases Research Center, Children's Hospital Boston, Enders Research Laboratories, 300 Longwood Ave., Boston, MA 02115, USA.
| | | |
Collapse
|
95
|
Lin DW, FitzGerald LM, Fu R, Kwon EM, Zheng SL, Kolb S, Wiklund F, Stattin P, Isaacs WB, Xu J, Ostrander EA, Feng Z, Grönberg H, Stanford JL. Genetic variants in the LEPR, CRY1, RNASEL, IL4, and ARVCF genes are prognostic markers of prostate cancer-specific mortality. Cancer Epidemiol Biomarkers Prev 2011; 20:1928-36. [PMID: 21846818 DOI: 10.1158/1055-9965.epi-11-0236] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Prostate cancer is the second leading cause of cancer-related deaths in men, accounting for more than 30,000 deaths annually. The purpose of this study was to test whether variation in selected candidate genes in biological pathways of interest for prostate cancer progression could help distinguish patients at higher risk for fatal prostate cancer. METHODS In this hypothesis-driven study, we genotyped 937 single nucleotide polymorphisms (SNPs) in 156 candidate genes in a population-based cohort of 1,309 prostate cancer patients. We identified 22 top-ranking SNPs (P ≤ 0.01, FDR ≤ 0.70) associated with prostate cancer-specific mortality (PCSM). A subsequent validation study was completed in an independent population-based cohort of 2,875 prostate cancer patients. RESULTS Five SNPs were validated (P ≤ 0.05) as being significantly associated with PCSM, one each in the LEPR, CRY1, RNASEL, IL4, and ARVCF genes. Compared with patients with 0 to 2 of the at-risk genotypes those with 4 to 5 at-risk genotypes had a 50% (95% CI, 1.2-1.9) higher risk of PCSM and risk increased with the number of at-risk genotypes carried (P(trend) = 0.001), adjusting for clinicopathologic factors known to influence prognosis. CONCLUSION Five genetic markers were validated to be associated with lethal prostate cancer. IMPACT This is the first population-based study to show that germline genetic variants provide prognostic information for prostate cancer-specific survival. The clinical utility of this five-SNP panel to stratify patients at higher risk for adverse outcomes should be evaluated.
Collapse
Affiliation(s)
- Daniel W Lin
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
XMRV Discovery and Prostate Cancer-Related Research. Adv Virol 2011; 2011:432837. [PMID: 22312343 PMCID: PMC3265305 DOI: 10.1155/2011/432837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/25/2011] [Indexed: 11/21/2022] Open
Abstract
Xenotropic murine leukemia virus-related virus (XMRV) was first reported in 2006 in a study of human prostate cancer patients with genetic variants of the antiviral enzyme, RNase L. Subsequent investigations in North America, Europe, Asia, and Africa have either observed or failed to detect XMRV in patients (prostate cancer, chronic fatigue syndrome-myalgic encephalomyelitis (CFS-ME), and immunosuppressed with respiratory tract infections) or normal, healthy, control individuals. The principal confounding factors are the near ubiquitous presence of mouse-derived reagents, antibodies and cells, and often XMRV itself, in laboratories. XMRV infects and replicates well in many human cell lines, but especially in certain prostate cancer cell lines. XMRV also traffics to prostate in a nonhuman primate model of infection. Here, we will review the discovery of XMRV and then focus on prostate cancer-related research involving this intriguing virus.
Collapse
|
97
|
Rusmevichientong A, Chow SA. Biology and pathophysiology of the new human retrovirus XMRV and its association with human disease. Immunol Res 2011; 48:27-39. [PMID: 20717743 DOI: 10.1007/s12026-010-8165-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Xenotropic murine leukemia virus-related virus (XMRV) is a new human retrovirus originally identified in prostate cancer patients with a deficiency in the antiviral enzyme RNase L. XMRV has been detected with varying frequencies in cases of prostate cancer and chronic fatigue syndrome (CFS), as well as in a small proportion of healthy individuals. An etiologic link between XMRV infection and human disease, however, has yet to be established. Here, we summarize existing knowledge regarding the characteristics of XMRV replication, association of XMRV with prostate cancer and CFS, and potential mechanisms of XMRV pathophysiology. We also highlight several areas, such as the establishment of standardized assays and the development of animal models, as future directions to advance our current understanding of XMRV and its relevance to human disease.
Collapse
Affiliation(s)
- Alice Rusmevichientong
- Department of Molecular and Medical Pharmacology, Molecular Biology Institute, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | | |
Collapse
|
98
|
New Virus Discovery in the 21st Century. Mol Microbiol 2011. [DOI: 10.1128/9781555816834.ch41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
99
|
Abstract
For decades, physicians and researchers have recognized that family history is a significant risk factor for prostate cancer. The identification of the genes responsible for inherited risk, however, proved difficult. With the sequencing of the human genome and the completion of the initial phases of the International HapMap Project, the tools are available to scan the entire genome and find genetic markers for disease. Since 2006, more than 30 inherited variants strongly associated with prostate cancer have been reported. As the inherited component of the disease is revealed, efforts are ongoing to translate genetic findings into the clinic.
Collapse
Affiliation(s)
- Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
100
|
Pakkanen S, Kujala PM, Ha N, Matikainen MP, Schleutker J, Tammela TL. Clinical and histopathological characteristics of familial prostate cancer in Finland. BJU Int 2011; 109:557-63. [DOI: 10.1111/j.1464-410x.2011.10198.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|