1
|
Acosta AM, Neil AJ, Hirsch MS. Primary mucinous adenocarcinoma of the seminal vesicle associated with intestinal metaplasia: a radiation-induced tumour? Histopathology 2021; 79:444-448. [PMID: 34096663 DOI: 10.1111/his.14425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Andres M Acosta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander J Neil
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Singh CK, Chhabra G, Ndiaye MA, Siddiqui IA, Panackal JE, Mintie CA, Ahmad N. Quercetin-Resveratrol Combination for Prostate Cancer Management in TRAMP Mice. Cancers (Basel) 2020; 12:E2141. [PMID: 32748838 PMCID: PMC7465013 DOI: 10.3390/cancers12082141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023] Open
Abstract
Prostate Cancer (PCa) is a leading cause of cancer-related morbidity and mortality in men. Therefore, novel mechanistically-driven approaches are needed for PCa management. Here, we determined the effects of grape antioxidants quercetin and/or resveratrol (60 and 600 mg/kg, respectively, in diet) against PCa in Transgenic Adenocarcinoma of Mouse Prostate (TRAMP)-model in prevention and intervention settings. We found resveratrol alone and in combination significantly inhibited prostate tumorigenesis in prevention setting, while the same was seen only in combination after intervention. The observed effects were associated with marked inhibition in proliferation, oxidative stress, and tumor survival markers, and induced apoptosis markers. Utilizing PCa PCR array analysis with prevention tumor tissues, we identified that quercetin-resveratrol modulates genes involved in promoter methylation, cell cycle, apoptosis, fatty acid metabolism, transcription factors, androgen response, PI3K/AKT and PTEN signaling. Ingenuity Pathway Analysis (IPA) identified IGF1 and BCL2 as central players in two gene networks. Functional annotation predicted increased apoptosis and inhibited cell viability/proliferation, hyperplasia, vasculogenesis, and angiogenesis with dual treatment. Furthermore, IPA predicted upstream inhibition of major PCa signaling VEGF, Ca2+, PI3K, CSF2, PTH). Based on PCR array, we identified decreased levels of EGFR, EGR3, and IL6, and increased levels of IGFBP7 and NKX3.1, overall supporting anti-PCa effects of quercetin-resveratrol.
Collapse
Affiliation(s)
- Chandra K. Singh
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Mary A. Ndiaye
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Imtiaz A. Siddiqui
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Jennifer E. Panackal
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Charlotte A. Mintie
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; (C.K.S.); (G.C.); (M.A.N.); (I.A.S.); (J.E.P.); (C.A.M.)
- William S. Middleton VA Medical Center, Madison, WI 53705, USA
| |
Collapse
|
3
|
Li T, Wang F, Dang Y, Dong J, Zhang Y, Zhang C, Liu P, Gao Y, Wang X, Yang S, Lu S. P21 and P27 promote tumorigenesis and progression via cell cycle acceleration in seminal vesicles of TRAMP mice. Int J Biol Sci 2019; 15:2198-2210. [PMID: 31592235 PMCID: PMC6775302 DOI: 10.7150/ijbs.35092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022] Open
Abstract
Transgenic adenocarcinoma mouse prostate (TRAMP) model is established to mimic human prostate cancer progression, where seminal vesicle lesions often occur and has been described as phyllodes-like epithelial-stromal tumors. However, the molecular mechanism regulating tumorigenesis and progression in seminal vesicles of TRAMP mice remains largely unknown. In this study, C57BL/6 TRAMP mice were found to have a significantly shorter lifespan than wild-type (WT) mice and all of the seminal vesicles were markedly increased in size and weight with age from 24 weeks exhibiting a clearly papillary-phyllode pattern, though no obvious difference was observed in multiple organs including heart, liver, spleen, lungs, kidneys, testicles and bone between TRAMP and WT mice, and less than 10% of TRAMP mice developed prostate tumors. Western blotting showed Cyclin (CCN) B1 and CCND1 were remarkably overexpressed in seminal vesicle tumors of TRAMP mice at 24 weeks of age and increased with age till the end of trial, which was confirmed by Immunohistochemistry (IHC). P21 and P27 were also significantly augmented, whereas P53 and phosphorylated P53 (p-P53) were constantly expressed in normal controls and P53 did not appear to be mutated. Not only cyclin-dependent kinase (CDK) 1 and phosphorylated forkhead box protein (FOX) O1 but also CDK4, CDK6 and phosphorylated retinoblastoma-associated protein (RB) had similar increase trends, so did epidermal growth factor receptor (EGFR), AKT serine/threonine kinase (AKT), and their respective phosphorylation levels. Signal transducer and activator of transcription (STAT) 3, p-STAT3, enhancer of zeste homolog 2 (EZH2) and EZH2 mediated trimethylation of histone H3 lysine 27 (H3K27me3) were considerably elevated, too. Taken together, this finding suggests P21 and P27 promote carcinogenesis and development in seminal vesicles of TRAMP mice via accelerating cell cycle progression, in which oncogenic transformation of P21 and P27 might be through regulation of EGFR-AKT signaling.
Collapse
Affiliation(s)
- Tonghui Li
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Fangfang Wang
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Yanmei Dang
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Jiajie Dong
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Yu Zhang
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Chi Zhang
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Ping Liu
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Yanhong Gao
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Xiaojun Wang
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| | - Sijun Yang
- ABSL-3 Laboratory at the Center for Animal Experiment and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, 430071, China
| | - Shan Lu
- School of Life Sciences, Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
4
|
Jachetti E, Rigoni A, Bongiovanni L, Arioli I, Botti L, Parenza M, Cancila V, Chiodoni C, Festinese F, Bellone M, Tardanico R, Tripodo C, Colombo MP. Imatinib Spares cKit-Expressing Prostate Neuroendocrine Tumors, whereas Kills Seminal Vesicle Epithelial-Stromal Tumors by Targeting PDGFR-β. Mol Cancer Ther 2016; 16:365-375. [PMID: 27980106 DOI: 10.1158/1535-7163.mct-16-0466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/31/2016] [Accepted: 11/19/2016] [Indexed: 11/16/2022]
Abstract
Prostate cancer is a leading cause of cancer-related death in males worldwide. Indeed, advanced and metastatic disease characterized by androgen resistance and often associated with neuroendocrine (NE) differentiation remains incurable. Using the spontaneous prostate cancer TRAMP model, we have shown that mast cells (MCs) support in vivo the growth of prostate adenocarcinoma, whereas their genetic or pharmacologic targeting favors prostate NE cancer arousal. Aiming at simultaneously targeting prostate NE tumor cells and MCs, both expressing the cKit tyrosine kinase receptor, we have tested the therapeutic effect of imatinib in TRAMP mice. Imatinib-treated TRAMP mice experience a partial benefit against prostate adenocarcinoma, because of inhibition of supportive MCs. However, they show an unexpected outgrowth of prostate NE tumors, likely because of defective signaling pathway downstream of cKit receptor. Also unexpected but very effective was the inhibition of epithelial-stromal tumors of the seminal vesicles achieved by imatinib treatment. These tumors normally arise in the seminal vesicles of TRAMP mice, independently of the degree of prostatic glandular lesions, and resemble phyllodes tumors found in human prostate and seminal vesicles, and in breast. In both mice and in patients, these tumors are negative for cKit but express PDGFR-β, another tyrosine kinase receptor specifically inhibited by imatinib. Our results imply a possible detrimental effect of imatinib in prostate cancer patients but suggest a promising therapeutic application of imatinib in the treatment of recurrent or metastatic phyllodes tumors. Mol Cancer Ther; 16(2); 365-75. ©2016 AACR.
Collapse
Affiliation(s)
- Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alice Rigoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Bongiovanni
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Ivano Arioli
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Botti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mariella Parenza
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Fabrizio Festinese
- Pharmacy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Bellone
- Division of Immunology, Transplantation and Infectious Diseases, Cellular Immunology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Regina Tardanico
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
5
|
Morphology and MMP-9, AR and IGFR-1 responses of the seminal vesicle in TRAMP mice model. Tissue Cell 2016; 48:217-23. [DOI: 10.1016/j.tice.2016.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 01/06/2023]
|
6
|
Mansfield DC, Kyula JN, Rosenfelder N, Chao-Chu J, Kramer-Marek G, Khan AA, Roulstone V, McLaughlin M, Melcher AA, Vile RG, Pandha HS, Khoo V, Harrington KJ. Oncolytic vaccinia virus as a vector for therapeutic sodium iodide symporter gene therapy in prostate cancer. Gene Ther 2016; 23:357-68. [PMID: 26814609 PMCID: PMC4827015 DOI: 10.1038/gt.2016.5] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/07/2015] [Accepted: 01/13/2016] [Indexed: 12/16/2022]
Abstract
Oncolytic strains of vaccinia virus are currently in clinical development with clear evidence of safety and promising signs of efficacy. Addition of therapeutic genes to the viral genome may increase the therapeutic efficacy of vaccinia. We evaluated the therapeutic potential of vaccinia virus expressing the sodium iodide symporter (NIS) in prostate cancer models, combining oncolysis, external beam radiotherapy and NIS-mediated radioiodide therapy. The NIS-expressing vaccinia virus (VV-NIS), GLV-1h153, was tested in in vitro analyzes of viral cell killing, combination with radiotherapy, NIS expression, cellular radioiodide uptake and apoptotic cell death in PC3, DU145, LNCaP and WPMY-1 human prostate cell lines. In vivo experiments were carried out in PC3 xenografts in CD1 nude mice to assess NIS expression and tumor radioiodide uptake. In addition, the therapeutic benefit of radioiodide treatment in combination with viral oncolysis and external beam radiotherapy was measured. In vitro viral cell killing of prostate cancers was dose- and time-dependent and was through apoptotic mechanisms. Importantly, combined virus therapy and iodizing radiation did not adversely affect oncolysis. NIS gene expression in infected cells was functional and mediated uptake of radioiodide both in vitro and in vivo. Therapy experiments with both xenograft and immunocompetent Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mouse models showed that the addition of radioiodide to VV-NIS-infected tumors was more effective than each single-agent therapy, restricting tumor growth and increasing survival. In conclusion, VV-NIS is effective in prostate cancer models. This treatment modality would be an attractive complement to existing clinical radiotherapy practice.
Collapse
Affiliation(s)
- D C Mansfield
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - J N Kyula
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - N Rosenfelder
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - J Chao-Chu
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - G Kramer-Marek
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - A A Khan
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - V Roulstone
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - M McLaughlin
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
| | - A A Melcher
- Leeds Institute of Cancer and Pathology, University of Leeds, St James's University Hospital, Leeds, UK
| | - R G Vile
- Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA
| | - H S Pandha
- Postgraduate Medical School, The University of Surrey, Guildford, UK
| | - V Khoo
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
- The Royal Marsden Hospital, London, UK
- University of Melbourne and Monash University, Victoria, Australia
| | - K J Harrington
- Divisions of Cancer Biology and Radiotherapy and Imaging, The Institute of Cancer Research, Chester Beatty Labs, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
7
|
Peek EM, Song W, Zhang H, Huang J, Chin AI. Loss of MyD88 leads to more aggressive TRAMP prostate cancer and influences tumor infiltrating lymphocytes. Prostate 2015; 75:463-73. [PMID: 25597486 DOI: 10.1002/pros.22932] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/22/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND The influence of pattern recognition receptor (PRR) signaling in the prostate tumor microenvironment remains unclear. Although there may be a role for PRR agonists as adjuvants to therapy, prior evidence suggests tumor promoting as well as tumor inhibiting mechanisms. The purpose of this study is to examine the role of the key Toll-like receptor (TLR) signaling adaptor protein myeloid differentiation primary response gene 88 (MyD88) in prostate cancer development. METHODS MyD88(-/-) mice in a C57Bl6 background were crossed with transgenic adenocarcinomas of the mouse prostate (TRAMP) mice to create MyD88(-/-) TRAMP(Tg+/-) animals, which were compared to MyD88(+/+) TRAMP(Tg+/-) animals and their non-transgenic counterparts at 30 weeks. Prostates were examined histologically, by immunohistochemistry and immunofluorescence staining, and by qPCR, to characterize tumor-infiltrating immune populations as well as activation of the downstream NF-κB pathway and androgen receptor (AR) expression. Splenocytes were examined for development of distinct immune cell populations. RESULTS Absence of MyD88 led to increased prostatic intraepithelial neoplasm (PIN) and areas of well-differentiated adenocarcinoma in TRAMP transgenic mice. Analysis of infiltrating immune populations revealed an increase in CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs), as evidenced by increased expression of prostatic arginase-1 and iNOS as well as the cytokine IL-10, and a deficiency in NK cells in prostates from MyD88(-/-) TRAMP(Tg+/-) compared to MyD88(+/+) TRAMP(Tg+/-) mice, whereas a decrease in splenocytic NK cell differentiation was observed in MyD88(-/-) mice. Prostate tumors revealed no significant differences in NF-κB or AR expression in MyD88(+/+) TRAMP(Tg+/-) compared to MyD88(-/-) TRAMP(Tg+/-) mice. CONCLUSIONS During prostate cancer development in the TRAMP model, MyD88 may play a role in limiting prostate tumorigenesis by altering tumor-infiltrating immune populations. This suggests that in the context of specific cancers, distinct PRRs and signaling pathways of innate immune signaling may influence the tumor microenvironment and represent a novel therapeutic strategy.
Collapse
|
8
|
Williams KA, Lee M, Hu Y, Andreas J, Patel SJ, Zhang S, Chines P, Elkahloun A, Chandrasekharappa S, Gutkind JS, Molinolo AA, Crawford NPS. A systems genetics approach identifies CXCL14, ITGAX, and LPCAT2 as novel aggressive prostate cancer susceptibility genes. PLoS Genet 2014; 10:e1004809. [PMID: 25411967 PMCID: PMC4238980 DOI: 10.1371/journal.pgen.1004809] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/06/2014] [Indexed: 11/23/2022] Open
Abstract
Although prostate cancer typically runs an indolent course, a subset of men develop aggressive, fatal forms of this disease. We hypothesize that germline variation modulates susceptibility to aggressive prostate cancer. The goal of this work is to identify susceptibility genes using the C57BL/6-Tg(TRAMP)8247Ng/J (TRAMP) mouse model of neuroendocrine prostate cancer. Quantitative trait locus (QTL) mapping was performed in transgene-positive (TRAMPxNOD/ShiLtJ) F2 intercross males (n = 228), which facilitated identification of 11 loci associated with aggressive disease development. Microarray data derived from 126 (TRAMPxNOD/ShiLtJ) F2 primary tumors were used to prioritize candidate genes within QTLs, with candidate genes deemed as being high priority when possessing both high levels of expression-trait correlation and a proximal expression QTL. This process enabled the identification of 35 aggressive prostate tumorigenesis candidate genes. The role of these genes in aggressive forms of human prostate cancer was investigated using two concurrent approaches. First, logistic regression analysis in two human prostate gene expression datasets revealed that expression levels of five genes (CXCL14, ITGAX, LPCAT2, RNASEH2A, and ZNF322) were positively correlated with aggressive prostate cancer and two genes (CCL19 and HIST1H1A) were protective for aggressive prostate cancer. Higher than average levels of expression of the five genes that were positively correlated with aggressive disease were consistently associated with patient outcome in both human prostate cancer tumor gene expression datasets. Second, three of these five genes (CXCL14, ITGAX, and LPCAT2) harbored polymorphisms associated with aggressive disease development in a human GWAS cohort consisting of 1,172 prostate cancer patients. This study is the first example of using a systems genetics approach to successfully identify novel susceptibility genes for aggressive prostate cancer. Such approaches will facilitate the identification of novel germline factors driving aggressive disease susceptibility and allow for new insights into these deadly forms of prostate cancer. Prostate cancer is a remarkably common disease, and in 2014 it is estimated that it will account for 27% of new cancer cases in men in the US. However, less than 13% those diagnosed will succumb to prostate cancer, with most men dying from unrelated causes. The tests used to identify men at risk of fatal prostate cancer are inaccurate, which leads to overtreatment, unnecessary patient suffering, and represents a significant public health burden. Many studies have shown that hereditary genetic variation significantly alters susceptibility to fatal prostate cancer, although the identities of genes responsible for this are mostly unknown. Here, we used a mouse model of prostate cancer to identify such genes. We introduced hereditary genetic variation into this mouse model through breeding, and used a genetic mapping technique to identify 35 genes associated with aggressive disease. The levels of three of these genes were consistently abnormal in human prostate cancers with a more aggressive disease course. Additionally, hereditary differences in these same three genes were associated with markers of fatal prostate cancer in men. This approach has given us unique insights into how hereditary variation influences fatal forms of prostate cancer.
Collapse
Affiliation(s)
- Kendra A. Williams
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Minnkyong Lee
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Ying Hu
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, Rockville, Maryland, United States of America
| | - Jonathan Andreas
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Shashank J. Patel
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Suiyuan Zhang
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Peter Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Abdel Elkahloun
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Settara Chandrasekharappa
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - J. Silvio Gutkind
- Oral & Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, United States of America
| | - Alfredo A. Molinolo
- Oral & Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, United States of America
| | - Nigel P. S. Crawford
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
9
|
Seminal Vesicle Intraepithelial Neoplasia Versus Basal Cell Hyperplasia in a Seminal Vesicle. Eur Urol 2014; 66:623-7. [DOI: 10.1016/j.eururo.2014.02.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/22/2014] [Indexed: 11/23/2022]
|
10
|
Espinoza I, Pochampally R, Xing F, Watabe K, Miele L. Notch signaling: targeting cancer stem cells and epithelial-to-mesenchymal transition. Onco Targets Ther 2013; 6:1249-59. [PMID: 24043949 PMCID: PMC3772757 DOI: 10.2147/ott.s36162] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Notch signaling is an evolutionarily conserved pathway involved in cell fate control during development, stem cell self-renewal, and postnatal tissue differentiation. Roles for Notch in carcinogenesis, the biology of cancer stem cells, tumor angiogenesis, and epithelial-to-mesenchymal transition (EMT) have been reported. This review describes the role of Notch in the "stemness" program in cancer cells and in metastases, together with a brief update on the Notch inhibitors currently under investigation in oncology. These agents may be useful in targeting cancer stem cells and to reverse the EMT process.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fei Xing
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kounosuke Watabe
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Microbiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lucio Miele
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
11
|
Jachetti E, Mazzoleni S, Grioni M, Ricupito A, Brambillasca C, Generoso L, Calcinotto A, Freschi M, Mondino A, Galli R, Bellone M. Prostate cancer stem cells are targets of both innate and adaptive immunity and elicit tumor-specific immune responses. Oncoimmunology 2013; 2:e24520. [PMID: 23762811 PMCID: PMC3667917 DOI: 10.4161/onci.24520] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/29/2013] [Accepted: 04/02/2013] [Indexed: 02/07/2023] Open
Abstract
According to the cancer stem cell (CSC) theory, therapies that do not target the CSC compartment have limited, if any, chances to eradicate established tumors. While cytotoxic T lymphocytes (CTLs) have the potential to recognize and kill single neoplastic cells within a tissue, whether CSCs can be targeted by the immune system during spontaneous or vaccination-elicited responses is poorly defined. Here, we provide experimental evidence showing that CSC lines established from the prostate of transgenic adenocarcinoma of the mouse prostate (TRAMP) mice expressed prostate cancer-associated antigens, MHC Class I and II molecules as well as ligands for natural killer (NK) cell receptors. Indeed, CSC were targets for both NK cell- and CTL-mediated cytotoxicity, both in vitro and in vivo. The administration of dendritic cells pulsed with irradiated CSCs induced a tumor-specific immune response that was more robust than that induced by dendritic cells pulsed with differentiated tumor cells, delayed tumor growth in mice challenged with prostate CSCs and caused tumor regression in TRAMP mice. Thus, CSC are targeted by both innate and adaptive immune responses and might be exploited for the design of novel immunotherapeutic approaches against cancer.
Collapse
Affiliation(s)
- Elena Jachetti
- Cellular Immunology Unit; PIBIC; Division of Immunology Transplantation and Infectious Disease; San Raffaele Scientific Institute; Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Jäämaa S, Laiho M. Maintenance of genomic integrity after DNA double strand breaks in the human prostate and seminal vesicle epithelium: the best and the worst. Mol Oncol 2012; 6:473-83. [PMID: 22762987 PMCID: PMC3439595 DOI: 10.1016/j.molonc.2012.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 01/23/2023] Open
Abstract
Prostate cancer is one of the most frequent cancer types in men, and its incidence is steadily increasing. On the other hand, primary seminal vesicle carcinomas are extremely rare with less than 60 cases reported worldwide. Therefore the difference in cancer incidence has been estimated to be more than a 100,000-fold. This is astonishing, as both tissues share similar epithelial structure and hormonal cues. Clearly, the two epithelia differ substantially in the maintenance of genomic integrity, possibly due to inherent differences in their DNA damage burden and DNA damage signaling. The DNA damage response evoked by DNA double strand breaks may be relevant, as their faulty repair has been implicated in the formation of common genomic rearrangements such as TMPRSS2-ERG fusions during prostate carcinogenesis. Here, we review DNA damaging processes of both tissues with an emphasis on inflammation and androgen signaling. We discuss how benign prostate and seminal vesicle epithelia respond to acute DNA damage, focusing on the canonical DNA double strand break-induced ATM-pathway, p53 and DNA damage induced checkpoints. We propose that the prostate might be more prone to the accumulation of genetic aberrations during epithelial regeneration than seminal vesicles due to a weaker ability to enforce DNA damage checkpoints.
Collapse
Affiliation(s)
- Sari Jäämaa
- Molecular Cancer Biology Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Marikki Laiho
- Molecular Cancer Biology Program, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
- The Sidney Kimmel Comprehensive Cancer Center, Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II, Room 444, Baltimore, MD 21231, USA
| |
Collapse
|
13
|
Jäämaa S, Sankila A, Rantanen V, Peltonen K, Järvinen PM, Af Hällström TM, Ruutu M, Taari K, Andersson LC, Laiho M. Contrasting DNA damage checkpoint responses in epithelium of the human seminal vesicle and prostate. Prostate 2012; 72:1060-70. [PMID: 22072329 DOI: 10.1002/pros.21509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 10/13/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND Prostate and seminal vesicle are two similar hormone responsive human organs that differ dramatically in their cancer incidence. DNA damage response (DDR) is required for maintenance of genomic integrity. METHODS In this study we investigated the DDR and cell cycle checkpoint activation of these organs using orthotopic cultures of human surgery-derived tissues and primary cultures of isolated prostate and seminal vesicle cells. RESULTS We find that the activation of ATM signaling pathway by ionizing radiation (IR) was comparable in both tissues. Previously, we have shown that the prostate secretory cells express low levels of histone variant H2AX and phosphorylated H2AX (γH2AX) after IR. Here we demonstrate that H2AX levels are low also in the secretory seminal vesicle cells suggesting that this is a common phenotype of postmitotic cells. We consequently established primary epithelial cell cultures from both organs to compare their DDR. Interestingly, contrary to human prostate epithelial cells (HPEC), primary seminal vesicle epithelial cells (HSVEC) displayed effective cell cycle checkpoints after IR and expressed higher levels of Wee1A checkpoint kinase. Furthermore, HSVEC but not HPEC cells were able to activate p53 and to induce p21 cell cycle inhibitor. DISCUSSION Our results show that during replication, the checkpoint enforcement is more proficient in the seminal vesicle than in the prostate epithelium cells. This indicates a more stringent enforcement of DDR in replicating seminal vesicle epithelial cells, and suggests that epithelial regeneration combined with sub-optimal checkpoint responses may contribute to high frequency of genetic lesions in the prostate epithelium.
Collapse
Affiliation(s)
- Sari Jäämaa
- Molecular Cancer Biology Program and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang L, Zhang J, Zhang Y, Nkhata K, Quealy E, Liao JD, Cleary MP, Lü J. Lobe-specific lineages of carcinogenesis in the transgenic adenocarcinoma of mouse prostate and their responses to chemopreventive selenium. Prostate 2011; 71:1429-40. [PMID: 21360561 DOI: 10.1002/pros.21360] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 01/19/2011] [Indexed: 11/09/2022]
Abstract
BACKGROUND The transgenic adenocarcinoma of mouse prostate (TRAMP) model is by far the most practical transgenic model for preclinical prostate cancer chemoprevention studies. It is critical to characterize the prostate lobe-specificity of lesion lineages to consolidate the advantages of this model and minimize its limitations for chemoprevention studies. METHODS We dissected dorsolateral (DLP), ventral (VP), and anterior prostate (AP) lobes, and macroscopic tumors from 90 male C57BL/6J TRAMP mice at 22-24 weeks of age (WOA) and analyzed lesions by histological, biochemical and proteomic approaches. To determine whether methylseleninic acid (MSeA) led to a deletion of initiated cells, we gave oral MSeA to TRAMP mice from 5 to 23 WOA or from 5 to 15 WOA and analyzed lesions at 23 WOA. RESULTS All tumors (n = 18) were T-antigen(+), synaptophysin (SYP)(+), androgen-receptor(-), and E-cadherin(-) poorly differentiated neuroendocrine carcinomas (NE-Ca). They were traceable most frequently to VP (66.7%) and rarely to DLP (11.1%) and AP (5.6%) with an estimated life-time incidence of 1 out of 3 mice. In DLP, epithelial lesions ranged from mild-to-severe atypical hyperplasia, with T-antigen(+), SYP(-), androgen-receptor(+), and E-cadherin(+). Proteomic profiling revealed many molecular differences between VP and DLP. In MSeA experiment, 6 out of 19 (31.5%) mice developed NE-Ca in the control group, only 2 in each MSeA group of 17-18 mice (11.1-11.8%) bore a detectable NE-Ca. CONCLUSION The C57BL/6J TRAMP mouse represents at least two lineages of prostate carcinogenesis. Chemoprevention studies should incorporate this knowledge for efficacy assessment and molecular target validations.
Collapse
Affiliation(s)
- Lei Wang
- Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Mack JT, Helke KL, Normand G, Green C, Townsend DM, Tew KD. ABCA2 transporter deficiency reduces incidence of TRAMP prostate tumor metastasis and cellular chemotactic migration. Cancer Lett 2010; 300:154-61. [PMID: 21041019 DOI: 10.1016/j.canlet.2010.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/15/2010] [Indexed: 10/18/2022]
Abstract
In order to study the effects of ATP-binding cassette transporter 2 (ABCA2) deficiency on the progression of prostate cancer, congenic Abca2 knockout (KO) mice were crossed to the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. ABCA2 expression was elevated in wild-type/TRAMP (WT/Tg) dorsal prostate, a region comprising the most aggressive tumors in this model, compared to non-transgenic WT mice. Primary prostate tumor progression was similar in KO/Tg and WT/Tg mice with respect to pathological score, prostate tumor growth, as calculated using MRI volumetry, and proliferative index, as determined by PCNA immunostaining. Vimentin, a marker of the epithelial-mesenchymal transition, was expressed at similar levels in prostate, but elevated in histologically normal seminal vesicles (SV) in KO/Tg mice (P < 0.02), concomitant with an increased SV volume (P < 0.01). These changes in the SV did not exacerbate the metastatic phenotype of this disease model; rather, KO/Tg mice aged 20-25 weeks had no detectable metastases while 38% of WT/Tg developed metastases to lung and/or lymph nodes. The absence of a metastatic phenotype in KO/Tg mice was reprised in stable ABCA2 knockdown (KD) cells where chemotactic, but not random, migration was impaired (P = 0.0004). Expression levels of sphingolipid biosynthetic enzymes were examined due to the established link of the transporter with sphingolipid homeostasis. Galactosylceramide synthase (GalCerS) mRNA levels were over 8-fold higher in KD cells (P = 0.001), while lactosylceramide synthase (LacCerS) and CTP:choline cytidylyltransferase (CCT) were significantly reduced (P < 0.0001 and 0.03, respectively). Overall, we demonstrate that ABCA2-deficiency inhibits prostate tumor metastasis in vivo and decreases chemotactic potential of cells, conceivably due to altered sphingolipid metabolism.
Collapse
Affiliation(s)
- Jody T Mack
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, 29425, United States
| | | | | | | | | | | |
Collapse
|
16
|
Zhang J, Wang L, Anderson LB, Witthuhn B, Xu Y, Lü J. Proteomic profiling of potential molecular targets of methyl-selenium compounds in the transgenic adenocarcinoma of mouse prostate model. Cancer Prev Res (Phila) 2010; 3:994-1006. [PMID: 20647336 DOI: 10.1158/1940-6207.capr-09-0261] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Because the Selenium (Se) and Vitamin E Cancer Prevention Trial (SELECT) failed to show the efficacy of selenomethionine for prostate cancer prevention, there is a critical need to identify safe and efficacious Se forms for future trials. We have recently shown significant preventive benefit of methylseleninic acid (MSeA) and Se-methylselenocysteine (MSeC) in the transgenic adenocarcinoma mouse prostate (TRAMP) model by oral administration. The present work applied iTRAQ proteomic approach to profile protein changes of the TRAMP prostate and to characterize their modulation by MSeA and MSeC to identify their potential molecular targets. Dorsolateral prostates from wild-type mice at 18 weeks of age and TRAMP mice treated with water (control), MSeA, or MSeC (3 mg Se/kg) from 8 to 18 weeks of age were pooled (9-10 mice per group) and subjected to protein extraction, followed by protein denaturation, reduction, and alkylation. After tryptic digestion, the peptides were labeled with iTRAQ reagents, mixed together, and analyzed by two-dimensional liquid chromatography/tandem mass spectrometry. Of 342 proteins identified with >95% confidence, the expression of 75 proteins was significantly different between TRAMP and wild-type mice. MSeA mainly affected proteins related to prostate functional differentiation, androgen receptor signaling, protein (mis)folding, and endoplasmic reticulum-stress responses, whereas MSeC affected proteins involved in phase II detoxification or cytoprotection, and in stromal cells. Although MSeA and MSeC are presumed precursors of methylselenol and were equally effective against the TRAMP model, their distinct affected protein profiles suggest biological differences in their molecular targets outweigh similarities.
Collapse
Affiliation(s)
- Jinhui Zhang
- The Hormel Institute, University of Minnesota, Austin, 55912, USA
| | | | | | | | | | | |
Collapse
|
17
|
Lopez-Barcons LA. Serially heterotransplanted human prostate tumours as an experimental model. J Cell Mol Med 2010; 14:1385-95. [PMID: 19874422 PMCID: PMC3829006 DOI: 10.1111/j.1582-4934.2009.00957.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 10/19/2009] [Indexed: 12/02/2022] Open
Abstract
* Introduction * Serially heterotransplanted human tumours in immunosuppressed mice: similarity to the tumour of origin - Cytological and histological analysis - Karyotype - Marker expression - Other PC markers - Tumour cell proliferation and frequency of mitosis - Vasculature - Stromal compartment - Heterotransplant hormone dependency - Androgen dependent - Partially androgen dependent - Androgen independent - Metastases * Conclusions Preclinical research on prostate cancer (PC) therapies uses several models to represent the human disease accurately. A common model uses patient prostate tumour biopsies to develop a cell line by serially passaging and subsequent implantation, in immunodeficient mice. An alternative model is direct implantation of patient prostate tumour biopsies into immunodeficient mice, followed by serial passage in vivo. The purpose of this review is to compile data from the more than 30 years of human PC serial heterotransplantation research. Serially heterotransplanted tumours are characterized by evaluating the histopathology of the resulting heterotransplants, including cellular differentiation, karyotype, marker expression, hormone sensitivity, cellular proliferation, metastatic potential and stromal and vascular components. These data are compared with the initial patient tumour specimen and, depending on available information, the patient's clinical outcome was compared with the heterotransplanted tumour. The heterotansplant model is a more accurate preclinical model than older generation serially passaged or genetic models to investigate current and newly developed androgen-deprivation agents, antitumour compounds, anti-angiogenic drugs and positron emission tomography radiotracers, as well as new therapeutic regimens for the treatment of PC.
Collapse
Affiliation(s)
- Lluis-A Lopez-Barcons
- Stanley S. Scott Cancer Center, Louisiana State University, Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|