51
|
Bernichtein S, Pigat N, Capiod T, Boutillon F, Verkarre V, Camparo P, Viltard M, Méjean A, Oudard S, Souberbielle JC, Friedlander G, Goffin V. High milk consumption does not affect prostate tumor progression in two mouse models of benign and neoplastic lesions. PLoS One 2015; 10:e0125423. [PMID: 25938513 PMCID: PMC4418739 DOI: 10.1371/journal.pone.0125423] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/23/2015] [Indexed: 01/25/2023] Open
Abstract
Epidemiological studies that have investigated whether dairy (mainly milk) diets are associated with prostate cancer risk have led to controversial conclusions. In addition, no existing study clearly evaluated the effects of dairy/milk diets on prostate tumor progression, which is clinically highly relevant in view of the millions of men presenting with prostate pathologies worldwide, including benign prostate hyperplasia (BPH) or high-grade prostatic intraepithelial neoplasia (HGPIN). We report here a unique interventional animal study to address this issue. We used two mouse models of fully penetrant genetically-induced prostate tumorigenesis that were investigated at the stages of benign hyperplasia (probasin-Prl mice, Pb-Prl) or pre-cancerous PIN lesions (KIMAP mice). Mice were fed high milk diets (skim or whole) for 15 to 27 weeks of time depending on the kinetics of prostate tumor development in each model. Prostate tumor progression was assessed by tissue histopathology examination, epithelial proliferation, stromal inflammation and fibrosis, tumor invasiveness potency and expression of various tumor markers relevant for each model (c-Fes, Gprc6a, activated Stat5 and p63). Our results show that high milk consumption (either skim or whole) did not promote progression of existing prostate tumors when assessed at early stages of tumorigenesis (hyperplasia and neoplasia). For some parameters, and depending on milk type, milk regimen could even exhibit slight protective effects towards prostate tumor progression by decreasing the expression of tumor-related markers like Ki-67 and Gprc6a. In conclusion, our study suggests that regular milk consumption should not be considered detrimental for patients presenting with early-stage prostate tumors.
Collapse
Affiliation(s)
- Sophie Bernichtein
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Natascha Pigat
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Thierry Capiod
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Florence Boutillon
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Virginie Verkarre
- Pathology Department, Hôpital Necker, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Paris, France
| | - Philippe Camparo
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Mélanie Viltard
- Institute for European Expertise in Physiology, Paris, France
| | - Arnaud Méjean
- Urology Department, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Paris, France
| | - Stéphane Oudard
- Medical Oncology Department, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Claude Souberbielle
- Inserm, U1151, Institut Necker Enfants Malades, Phosphate Homeostasis Laboratory, Paris, France
- Physiology Department, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Paris, France
| | - Gérard Friedlander
- Inserm, U1151, Institut Necker Enfants Malades, Phosphate Homeostasis Laboratory, Paris, France
- Physiology Department, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Paris, France
| | - Vincent Goffin
- Inserm, U1151, Institut Necker Enfants Malades, PRL/GH Pathophysiology Laboratory, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- * E-mail:
| |
Collapse
|
52
|
Bernichtein S, Pigat N, Camparo P, Latil A, Viltard M, Friedlander G, Goffin V. Anti-inflammatory properties of Lipidosterolic extract of Serenoa repens (Permixon®) in a mouse model of prostate hyperplasia. Prostate 2015; 75:706-22. [PMID: 25683150 DOI: 10.1002/pros.22953] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Permixon®, the hexanic lipidosterolic extract of saw palmetto Serenoa repens (LSESr), has shown properties that highlight its benefit in the management of benign prostate hyperplasia (BPH). To address its actual anti-inflammatory potency, we used a unique pro-inflammatory mouse model of prostate hyperplasia involving prostate-specific over-expression of prolactin transgene (Pb-Prl). METHODS Six month-old Pb-Prl males were administered with Permixon® per os at the daily dose of 100 mg/kg for 28 days. Body and prostate weights were measured weekly and at sacrifice, respectively. Prostate histology was carefully assessed by a pathologist and detailed quantifications of epithelial and stromal compartments were performed using image analysis software. Luminal cell proliferation index was determined using Ki-67 immunostaining, and apoptosis using Bax/Bcl2 mRNA ratio. Tissue inflammation and fibrosis were assessed by histological analyses then quantified using CD45 immunostaining and picrosirius staining, respectively. Expression profiling of selected pro-inflammatory cytokines, chemokines, and chemokine receptors was performed by quantitative RT-PCR. RESULTS In this model, Permixon® significantly decreased tissue weight and proliferation index specifically in the ventral lobe. Although treatment had no noticeable effect on epithelial histology of any lobe, it markedly reduced the histological hallmarks of inflammation in all lobes. This was confirmed by the global down-regulation of prostate pro-inflammatory cytokine profile, with significant reduction of CCR7, CXCL6, IL-6, and IL-17 expression. CONCLUSIONS In this mouse model of prostate hyperplasia, Permixon® exerted potent anti-inflammatory properties in the whole prostate while anti-androgenic effects were lobe-specific, suggesting that distinct LSESr components may be involved in these effects. Our results support the beneficial role of Permixon® treatment for BPH. The relevance of CCR7, CXCL6, IL-6, and IL-17 as potential biomarkers to follow up BPH inflammatory status needs to be assessed.
Collapse
|
53
|
Abstract
When the National Institutes of Health Mouse Models of Human Cancer Consortium initiated the Prostate Steering Committee 15 years ago, there were no genetically engineered mouse (GEM) models of prostate cancer (PCa). Today, a PubMed search for "prostate cancer mouse model" yields 3,200 publications and this list continues to grow. The first generation of GEM utilized the newly discovered and characterized probasin promoter driving viral oncogenes such as Simian virus 40 large T antigen to yield the LADY and TRAMP models. As the PCa research field has matured, the second generation of models has incorporated the single and multiple molecular changes observed in human disease, such as loss of PTEN and overexpression of Myc. Application of these models has revealed that mice are particularly resistant to developing invasive PCa, and once they achieve invasive disease, the PCa rarely resembles human disease. Nevertheless, these models and their application have provided vital information on human PCa progression. The aim of this review is to provide a brief primer on mouse and human prostate histology and pathology, provide descriptions of mouse models, as well as attempt to answer the age old question: Which GEM model of PCa is the best for my research question?
Collapse
|
54
|
Han JH, Park JH, Kim BY, Chang SN, Kim TH, Park JH, Kim DJ. Decreased expression of Toll-like receptor 4 and 5 during progression of prostate transformation in transgenic adenocarcinoma of mouse prostate mice. J Vet Sci 2015; 16:281-7. [PMID: 25797291 PMCID: PMC4588013 DOI: 10.4142/jvs.2015.16.3.281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 03/07/2015] [Indexed: 11/25/2022] Open
Abstract
Chronic inflammation has been considered an important risk factor for development of prostate cancer. Toll-like receptors (TLRs) recognize microbial moieties or endogenous molecules and play an important role in the triggering and promotion of inflammation. In this study, we examined whether expression of TLR4 and TLR5 was associated with progression of prostate transformation in the transgenic adenocarcinoma of mouse prostate (TRAMP) model. The expression of TLR4 and TLR5 was evaluated by immunohistochemisty in formalin-fixed paraffin-embedded prostate tissue from wild-type (WT) and TRAMP mice. Normal prostate tissue from WT mice showed strong expression of TLR4 and TLR5. However, TLR4 expression in the prostate tissue from TRAMP mice gradually decreased as pathologic grade became more aggressive. TLR5 expression in the prostate tissue from TRAMP mice also decreased in low-grade prostate intraepithelial neoplasia (PIN), high-grade PIN and poorly differentiated adenocarcinoma. Overall, our results suggest that decreased expression of TLR4 and TLR5 may contribute to prostate tumorigenesis.
Collapse
Affiliation(s)
- Ju-Hee Han
- Laboratory Animal Medicine, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
55
|
Linn DE, Bronson RT, Li Z. Genetic interaction between Tmprss2-ERG gene fusion and Nkx3.1-loss does not enhance prostate tumorigenesis in mouse models. PLoS One 2015; 10:e0120628. [PMID: 25780911 PMCID: PMC4364018 DOI: 10.1371/journal.pone.0120628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/25/2015] [Indexed: 11/19/2022] Open
Abstract
Gene fusions involving ETS family transcription factors (mainly TMPRSS2-ERG and TMPRSS2-ETV1 fusions) have been found in ~50% of human prostate cancer cases. Although expression of TMPRSS2-ERG or TMPRSS2-ETV1 fusion alone is insufficient to initiate prostate tumorigenesis, they appear to sensitize prostate epithelial cells for cooperation with additional oncogenic mutations to drive frank prostate adenocarcinoma. To search for such ETS-cooperating oncogenic events, we focused on a well-studied prostate tumor suppressor NKX3.1, as loss of NKX3.1 is another common genetic alteration in human prostate cancer. Previous studies have shown that deletions at 8p21 (harboring NKX3.1) and 21q22 (resulting in TMPRSS2-ERG fusion) were both present in a subtype of prostate cancer cases, and that ERG can lead to epigenetic silencing of NKX3.1 in prostate cancer cells, whereas NKX3.1 can in turn negatively regulate TMPRSS2-ERG fusion expression via suppression of the TMPRSS2 promoter activity. We recently generated knockin mouse models for TMPRSS2-ERG and TMPRSS2-ETV1 fusions, utilizing the endogenous Tmprss2 promoter. We crossed these knockin models to an Nkx3.1 knockout mouse model. In Tmprss2-ERG;Nkx3.1+/- (or -/-) male mice, although we observed a slight but significant upregulation of Tmprss2-ERG fusion expression upon Nkx3.1 loss, we did not detect any significant cooperation between these two genetic events to enhance prostate tumorigenesis in vivo. Furthermore, retrospective analysis of a previously published human prostate cancer dataset revealed that within ERG-overexpressing prostate cancer cases, NKX3.1 loss or deletion did not predict biochemical relapse after radical prostatectomy. Collectively, these data suggest that although TMPRSS2-ERG fusion and loss of NKX3.1 are among the most common mutational events found in prostate cancer, and although each of them can sensitize prostate epithelial cells for cooperating with other oncogenic events, these two events themselves do not appear to cooperate at a significant level in vivo to enhance prostate tumorigenesis.
Collapse
Affiliation(s)
- Douglas E. Linn
- Division of Genetics, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States of America
| | - Roderick T. Bronson
- Rodent Histopathology, Harvard Medical School, Boston, Massachusetts 02115, United States of America
| | - Zhe Li
- Division of Genetics, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States of America
- * E-mail:
| |
Collapse
|
56
|
Higgins J, Brogley M, Palanisamy N, Mehra R, Ittmann MM, Li JZ, Tomlins SA, Robins DM. Interaction of the Androgen Receptor, ETV1, and PTEN Pathways in Mouse Prostate Varies with Pathological Stage and Predicts Cancer Progression. Discov Oncol 2015; 6:67-86. [PMID: 25631336 DOI: 10.1007/s12672-014-0215-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022] Open
Abstract
To examine the impact of common somatic mutations in prostate cancer (PCa) on androgen receptor (AR) signaling, mouse models were designed to perturb sequentially the AR, ETV1, and PTEN pathways. Mice with "humanized" AR (hAR) alleles that modified AR transcriptional strength by varying polyglutamine tract (Q-tract) length were crossed with mice expressing a prostate-specific, AR-responsive ETV1 transgene (ETV1(Tg)). While hAR allele did not grossly affect ETV1-induced neoplasia, ETV1 strongly antagonized global AR regulation and repressed critical androgen-induced differentiation and tumor suppressor genes, such as Nkx3-1 and Hoxb13. When Pten was varied to determine its impact on disease progression, mice lacking one Pten allele (Pten(+/-) ) developed more frequent prostatic intraepithelial neoplasia (PIN). Yet, only those with the ETV1 transgene progressed to invasive adenocarcinoma. Furthermore, progression was more frequent with the short Q-tract (stronger) AR, suggesting that the AR, ETV1, and PTEN pathways cooperate in aggressive disease. On the Pten(+/-) background, ETV1 had markedly less effect on AR target genes. However, a strong inflammatory gene expression signature, notably upregulation of Cxcl16, was induced by ETV1. Comparison of mouse and human patient data stratified by the presence of E26 transformation-specific ETS fusion genes highlighted additional factors, some not previously associated with prostate cancer but for which targeted therapies are in development for other diseases. In sum, concerted use of these mouse models illuminates the complex interplay of AR, ETV1, and PTEN pathways in pre-cancerous neoplasia and early tumorigenesis, disease stages difficult to analyze in man.
Collapse
Affiliation(s)
- Jake Higgins
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Tumor-suppressive activity of Lunatic Fringe in prostate through differential modulation of Notch receptor activation. Neoplasia 2014; 16:158-67. [PMID: 24709423 DOI: 10.1593/neo.131870] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/06/2014] [Accepted: 01/17/2014] [Indexed: 01/10/2023] Open
Abstract
Elevated Notch ligand and receptor expression has been associated with aggressive forms of prostate cancer, suggesting a role for Notch signaling in regulation of prostate tumor initiation and progression. Here, we report a critical role for Lunatic Fringe (Lfng), which encodes an O-fucosylpeptide 3-ß-N-acetylglucosaminyltransferase known to modify epidermal growth factor repeats of Notch receptor proteins, in regulation of prostate epithelial differentiation and proliferation, as well as in prostate tumor suppression. Deletion of Lfng in mice caused altered Notch activation in the prostate, associated with elevated accumulation of Notch1, Notch2, and Notch4 intracellular domains, decreased levels of the putative Notch3 intracellular fragment, as well as increased expression of Hes1, Hes5, and Hey2. Loss of Lfng resulted in expansion of the basal layer, increased proliferation of both luminal and basal cells, and ultimately, prostatic intraepithelial neoplasia. The Lfng-null prostate showed down-regulation of prostatic tumor suppressor gene NKX3.1 and increased androgen receptor expression. Interestingly, expression of LFNG and NKX3.1 were positively correlated in publically available human prostate cancer data sets. Knockdown of LFNG in DU-145 prostate cancer cells led to expansion of CD44(+)CD24(-) and CD49f(+)CD24(-) stem/progenitor-like cell population associated with enhanced prostatosphere-forming capacity. Taken together, these data revealed a tumor-suppressive role for Lfng in the prostate through differential regulation of Notch signaling.
Collapse
|
58
|
Burcham GN, Cresswell GM, Snyder PW, Chen L, Liu X, Crist SA, Henry MD, Ratliff TL. Impact of prostate inflammation on lesion development in the POET3(+)Pten(+/-) mouse model of prostate carcinogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3176-91. [PMID: 25455686 DOI: 10.1016/j.ajpath.2014.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 07/14/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022]
Abstract
Evidence linking prostatitis and prostate cancer development is contradictory. To study this link, the POET3 mouse, an inducible model of prostatitis, was crossed with a Pten-loss model of prostate cancer (Pten(+/-)) containing the ROSA26 luciferase allele to monitor prostate size. Prostatitis was induced, and prostate bioluminescence was tracked over 12 months, with lesion development, inflammation, and cytokine expression analyzed at 4, 8, and 12 months and compared with mice without induction of prostatitis. Acute prostatitis led to more proliferative epithelium and enhanced bioluminescence. However, 4 months after initiation of prostatitis, mice with induced inflammation had lower grade pre-neoplastic lesions. A trend existed toward greater development of carcinoma 12 months after induction of inflammation, including one of two mice with carcinoma developing perineural invasion. Two of 18 mice at the later time points developed lesions with similarities to proliferative inflammatory atrophy, including one mouse with associated carcinoma. Pten(+/-) mice developed spontaneous inflammation, and prostatitis was similar among groups of mice at 8 and 12 months. Analyzed as one cohort, lesion number and grade were positively correlated with prostatitis. Specifically, amounts of CD11b(+)Gr1(+) cells were correlated with lesion development. These results support the hypothesis that myeloid-based inflammation is associated with lesion development in the murine prostate, and previous bouts of CD8-driven prostatitis may promote invasion in the Pten(+/-) model of cancer.
Collapse
Affiliation(s)
- Grant N Burcham
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; Heeke Animal Disease Diagnostic Laboratory, Southern Indiana Purdue Agricultural Center, Dubois, Indiana
| | - Gregory M Cresswell
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Paul W Snyder
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; Purdue University Center for Cancer Research, West Lafayette, Indiana
| | - Long Chen
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana
| | - Xiaoqi Liu
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana; Purdue University Center for Cancer Research, West Lafayette, Indiana
| | - Scott A Crist
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; Purdue University Center for Cancer Research, West Lafayette, Indiana
| | - Michael D Henry
- Department of Physiology and Biophysics and Holden Comprehensive Cancer Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; Purdue University Center for Cancer Research, West Lafayette, Indiana.
| |
Collapse
|
59
|
Xing C, Ci X, Sun X, Fu X, Zhang Z, Dong EN, Hao ZZ, Dong JT. Klf5 deletion promotes Pten deletion-initiated luminal-type mouse prostate tumors through multiple oncogenic signaling pathways. Neoplasia 2014; 16:883-99. [PMID: 25425963 PMCID: PMC4240924 DOI: 10.1016/j.neo.2014.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/13/2014] [Accepted: 09/22/2014] [Indexed: 01/18/2023] Open
Abstract
Krüppel-like factor 5 (KLF5) regulates multiple biologic processes. Its function in tumorigenesis appears contradictory though, showing both tumor suppressor and tumor promoting activities. In this study, we examined whether and how Klf5 functions in prostatic tumorigenesis using mice with prostate-specific deletion of Klf5 and phosphatase and tensin homolog (Pten), both of which are frequently inactivated in human prostate cancer. Histologic analysis demonstrated that when one Pten allele was deleted, which causes mouse prostatic intraepithelial neoplasia (mPIN), Klf5 deletion accelerated the emergence and progression of mPIN. When both Pten alleles were deleted, which causes prostate cancer, Klf5 deletion promoted tumor growth, increased cell proliferation, and caused more severe morphologic and molecular alterations. Homozygous deletion of Klf5 was more effective than hemizygous deletion. Unexpectedly, while Pten deletion alone expanded basal cell population in a tumor as reported, Klf5 deletion in the Pten-null background clearly reduced basal cell population while expanding luminal cell population. Global gene expression profiling, pathway analysis, and experimental validation indicate that multiple mechanisms could mediate the tumor-promoting effect of Klf5 deletion, including the up-regulation of epidermal growth factor and its downstream signaling molecules AKT and ERK and the inactivation of the p15 cell cycle inhibitor. KLF5 also appears to cooperate with several transcription factors, including CREB1, Sp1, Myc, ER and AR, to regulate gene expression. These findings validate the tumor suppressor function of KLF5. They also yield a mouse model that shares two common genetic alterations with human prostate cancer—mutation/deletion of Pten and deletion of Klf5.
Collapse
Affiliation(s)
- Changsheng Xing
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China ; Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Xinpei Ci
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China ; Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaodong Sun
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA ; Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhiqian Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric N Dong
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhao-Zhe Hao
- Department of Biology, University of Oklahoma, Norman, OK, USA
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China ; Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
60
|
Qin J, Lee HJ, Wu SP, Lin SC, Lanz RB, Creighton CJ, DeMayo FJ, Tsai SY, Tsai MJ. Androgen deprivation-induced NCoA2 promotes metastatic and castration-resistant prostate cancer. J Clin Invest 2014; 124:5013-26. [PMID: 25295534 DOI: 10.1172/jci76412] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/04/2014] [Indexed: 01/05/2023] Open
Abstract
A major clinical hurdle for the management of advanced prostate cancer (PCa) in patients is the resistance of tumors to androgen deprivation therapy (ADT) and their subsequent development into castration-resistant prostate cancer (CRPC). While recent studies have identified potential pathways involved in CRPC development, the drivers of CRPC remain largely undefined. Here we determined that nuclear receptor coactivator 2 (NCoA2, also known as SRC-2), which is frequently amplified or overexpressed in patients with metastatic PCa, mediates development of CRPC. In a murine model, overexpression of NCoA2 in the prostate epithelium resulted in neoplasia and, in combination with Pten deletion, promoted the development of metastasis-prone cancer. Moreover, depletion of NCoA2 in PTEN-deficient mice prevented the development of CRPC. In human androgen-sensitive prostate cancer cells, androgen signaling suppressed NCoA2 expression, and NCoA2 overexpression in murine prostate tumors resulted in hyperactivation of PI3K/AKT and MAPK signaling, promoting tumor malignance. Analysis of PCa patient samples revealed a strong correlation among NCoA2-mediated signaling, disease progression, and PCa recurrence. Taken together, our findings indicate that androgen deprivation induces NCoA2, which in turn mediates activation of PI3K signaling and promotes PCa metastasis and CRPC development. Moreover, these results suggest that the inhibition of NCoA2 has potential for PCa therapy.
Collapse
|
61
|
Chang SN, Han J, Abdelkader TS, Kim TH, Lee JM, Song J, Kim KS, Park JH, Park JH. High animal fat intake enhances prostate cancer progression and reduces glutathione peroxidase 3 expression in early stages of TRAMP mice. Prostate 2014; 74:1266-77. [PMID: 25053105 DOI: 10.1002/pros.22843] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 06/03/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Prostate cancer is the most frequently diagnosed cancer in Western men, and more men have been diagnosed at younger ages in recent years. A high-fat Western-style diet is a known risk factor for prostate cancer and increases oxidative stress. METHODS We evaluated the association between dietary animal fat and expression of antioxidant enzymes, particularly glutathione peroxidase 3 (GPx3), in the early stages of transgenic adenocarcinoma of the mouse prostate (TRAMP) mice. Six-week-old male nontransgenic and TRAMP mice were placed on high animal fat (45% Kcal fat) or control (10% Kcal fat) diets and sacrificed after 5 or 10 weeks. RESULTS The histopathological score increased with age and high-fat diet consumption. The histopathological scores in dorsal and lateral lobes increased in the 10-week high-fat diet group (6.2±0.2 and 6.2±0.4, respectively) versus the 10-week control diet group (5.3±0.3 and 5.2±0.2, respectively). GPx3 decreased both at the mRNA and protein levels in mouse prostate. GPx3 mRNA expression decreased (∼36.27% and ∼23.91%, respectively) in the anterior and dorsolateral prostate of TRAMP mice fed a high-fat diet compared to TRAMP mice fed a control diet. Cholesterol treatment increased PC-3 human prostate cancer cell proliferation, decreased GPx3 mRNA and protein levels, and increased H2 O2 levels in culture medium. Moreover, increasing GPx3 mRNA expression by troglitazone in PC-3 cells decreased cell proliferation and lowered H2 O2 levels. CONCLUSIONS Dietary fat enhances prostate cancer progression, possibly by suppressing GPx3 expression and increasing proliferation of prostate intraepithelial neoplasia (PIN) epithelial cells.
Collapse
Affiliation(s)
- Seo-Na Chang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
PSGR promotes prostatic intraepithelial neoplasia and prostate cancer xenograft growth through NF-κB. Oncogenesis 2014; 3:e114. [PMID: 25111863 PMCID: PMC5189964 DOI: 10.1038/oncsis.2014.29] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/22/2014] [Accepted: 07/02/2014] [Indexed: 12/16/2022] Open
Abstract
Prostate-specific G-protein-coupled receptor (PSGR), a member of the olfactory subfamily of G-protein-coupled receptors, is specifically expressed in human prostate tissue and overexpressed in prostate cancer (PCa). This expression pattern suggests a possible role in PCa initiation and progression. We developed a PSGR transgenic mouse model driven by a probasin promoter and investigated the role of PSGR in prostate malignancy. Overexpression of PSGR induced a chronic inflammatory response that ultimately gave rise to premalignant mouse prostate intraepithelial neoplasia lesions in later stages of life. PSGR-overexpressing LnCaP cells in prostate xenografts formed larger tumors compared with normal LnCaP cancer cells, suggesting a role of PSGR in the promotion of tumor development. Furthermore, we identified nuclear factor-κB (NF-κB) or RELA as a key downstream target activated by PSGR signaling. We also show that this regulation was mediated in part by the phosphatidylinositol-3-kinase/Akt (PI3K/AKT) pathway, highlighting a collaborative role between PI3K/AKT and NF-κB during tumor inflammation downstream of PSGR in the initial phases of prostate disease.Oncogenesis (2014) 3, e114; doi:10.1038/oncsis.2014.29; published online 11 August 2014.
Collapse
|
63
|
Kwon OJ, Valdez J, Zhang L, Zhang B, Wei X, Su Q, Ittmann MM, Creighton CJ, Xin L. Increased Notch signalling inhibits anoikis and stimulates proliferation of prostate luminal epithelial cells. Nat Commun 2014; 5:4416. [PMID: 25048699 PMCID: PMC4167399 DOI: 10.1038/ncomms5416] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/16/2014] [Indexed: 02/07/2023] Open
Abstract
The prostate epithelial lineage hierarchy remains inadequately defined. Recent lineage-tracing studies have implied the existence of prostate luminal epithelial progenitors with extensive regenerative capacity. However, this capacity has not been demonstrated in prostate stem cell activity assays, probably owing to the strong susceptibility of luminal progenitors to anoikis. Here we show that constitutive expression of Notch1 intracellular domain impairs secretory function of mouse prostate luminal cells, suppresses anoikis of luminal epithelial cells by augmenting NF-κB activity independent of Hes1, stimulates luminal cell proliferation by potentiating PI3K-AKT signalling, and rescues the capacities of the putative prostate luminal progenitors for unipotent differentiation in vivo and short-term self-renewal in vitro. Epithelial cell autonomous AR signalling is dispensable for the Notch-mediated effects. As Notch activity is increased in prostate cancers, and anoikis resistance is a hallmark for metastatic cancer cells, this study suggests a pro-metastatic function of Notch signalling during prostate cancer progression.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Joseph Valdez
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Li Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Xing Wei
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Qingtai Su
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine
- Dan L. Duncan Cancer Center, Baylor College of Medicine
- Michael E. DeBakey Department of Veterans Affairs Medical Center
| | | | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine
- Department of Pathology and Immunology, Baylor College of Medicine
- Dan L. Duncan Cancer Center, Baylor College of Medicine
| |
Collapse
|
64
|
Sun X, Fu X, Li J, Xing C, Frierson HF, Wu H, Ding X, Ju T, Cummings RD, Dong JT. Deletion of atbf1/zfhx3 in mouse prostate causes neoplastic lesions, likely by attenuation of membrane and secretory proteins and multiple signaling pathways. Neoplasia 2014; 16:377-89. [PMID: 24934715 PMCID: PMC4198693 DOI: 10.1016/j.neo.2014.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/30/2014] [Accepted: 05/06/2014] [Indexed: 01/14/2023] Open
Abstract
The ATBF1/ZFHX3 gene at 16q22 is the second most frequently mutated gene in human prostate cancer and has reduced expression or mislocalization in several types of human tumors. Nonetheless, the hypothesis that ATBF1 has a tumor suppressor function in prostate cancer has not been tested. In this study, we examined the role of ATBF1 in prostatic carcinogenesis by specifically deleting Atbf1 in mouse prostatic epithelial cells. We also examined the effect of Atbf1 deletion on gene expression and signaling pathways in mouse prostates. Histopathologic analyses showed that Atbf1 deficiency caused hyperplasia and mouse prostatic intraepithelial neoplasia (mPIN) primarily in the dorsal prostate but also in other lobes. Hemizygous deletion of Atbf1 also increased the development of hyperplasia and mPIN, indicating a haploinsufficiency of Atbf1. The mPIN lesions expressed luminal cell markers and harbored molecular changes similar to those in human PIN and prostate cancer, including weaker expression of basal cell marker cytokeratin 5 (Ck5), cell adhesion protein E-cadherin, and the smooth muscle layer marker Sma; elevated expression of the oncoproteins phospho-Erk1/2, phospho-Akt and Muc1; and aberrant protein glycosylation. Gene expression profiling revealed a large number of genes that were dysregulated by Atbf1 deletion, particularly those that encode for secretory and cell membrane proteins. The four signaling networks that were most affected by Atbf1 deletion included those centered on Erk1/2 and IGF1, Akt and FSH, NF-κB and progesterone and β-estradiol. These findings provide in vivo evidence that ATBF1 is a tumor suppressor in the prostate, suggest that loss of Atbf1 contributes to tumorigenesis by dysregulating membrane and secretory proteins and multiple signaling pathways, and provide a new animal model for prostate cancer.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322; Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jie Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Changsheng Xing
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, VA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322
| | - Xiaokun Ding
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | - Tongzhong Ju
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | | | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322.
| |
Collapse
|
65
|
González-Billalabeitia E, Seitzer N, Song SJ, Song MS, Patnaik A, Liu XS, Epping MT, Papa A, Hobbs RM, Chen M, Lunardi A, Ng C, Webster KA, Signoretti S, Loda M, Asara JM, Nardella C, Clohessy JG, Cantley LC, Pandolfi PP. Vulnerabilities of PTEN-TP53-deficient prostate cancers to compound PARP-PI3K inhibition. Cancer Discov 2014; 4:896-904. [PMID: 24866151 DOI: 10.1158/2159-8290.cd-13-0230] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Prostate cancer is the most prevalent cancer in males, and treatment options are limited for advanced forms of the disease. Loss of the PTEN and TP53 tumor suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often observed in advanced prostate cancer. Here, we show that PARP inhibition triggers a p53-dependent cellular senescence in a PTEN-deficient setting in the prostate. Surprisingly, we also find that PARP-induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and p53. We further show that superactivation of the prosurvival PI3K-AKT signaling pathway limits the efficacy of a PARP single-agent treatment, and that PARP and PI3K inhibitors effectively synergize to suppress tumorigenesis in human prostate cancer cell lines and in a Pten/Trp53-deficient mouse model of advanced prostate cancer. Our findings, therefore, identify a combinatorial treatment with PARP and PI3K inhibitors as an effective option for PTEN-deficient prostate cancer. SIGNIFICANCE The paucity of therapeutic options in advanced prostate cancer displays an urgent need for the preclinical assessment of novel therapeutic strategies. We identified differential therapeutic vulnerabilities that emerge upon the loss of both PTEN and p53, and observed that combined inhibition of PARP and PI3K provides increased efficacy in hormone-insensitive advanced prostate cancer.
Collapse
Affiliation(s)
- Enrique González-Billalabeitia
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of On leave of absence: Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Nina Seitzer
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Su Jung Song
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Min Sup Song
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Akash Patnaik
- Hematology/Oncology and Signal Transduction, Department of Medicine; Department of Systems Biology, Harvard Medical School
| | - Xue-Song Liu
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Mirjam T Epping
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Antonella Papa
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Robin M Hobbs
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Andrea Lunardi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Christopher Ng
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Kaitlyn A Webster
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| | - Sabina Signoretti
- Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
| | - Massimo Loda
- Department of Pathology, Brigham and Women's Hospital; Center for Molecular Oncologic Pathology; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - John M Asara
- Department of Systems Biology, Harvard Medical School
| | - Caterina Nardella
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center
| | - Lewis C Cantley
- Signal Transduction, Department of Medicine; Department of Systems Biology, Harvard Medical School
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Medicine; Divisions of
| |
Collapse
|
66
|
Lin SJ, Lee SO, Lee YF, Miyamoto H, Yang DR, Li G, Chang C. TR4 nuclear receptor functions as a tumor suppressor for prostate tumorigenesis via modulation of DNA damage/repair system. Carcinogenesis 2014; 35:1399-406. [PMID: 24583925 DOI: 10.1093/carcin/bgu052] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Testicular nuclear receptor 4 (TR4), a member of the nuclear receptor superfamily, plays important roles in metabolism, fertility and aging. The linkage of TR4 functions in cancer progression, however, remains unclear. Using three different mouse models, we found TR4 could prevent or delay prostate cancer (PCa)/prostatic intraepithelial neoplasia development. Knocking down TR4 in human RWPE1 and mouse mPrE normal prostate cells promoted tumorigenesis under carcinogen challenge, suggesting TR4 may play a suppressor role in PCa initiation. Mechanism dissection in both in vitro cell lines and in vivo mice studies found that knocking down TR4 led to increased DNA damage with altered DNA repair system that involved the modulation of ATM expression at the transcriptional level, and addition of ATM partially interrupted the TR4 small interfering RNA-induced tumorigenesis in cell transformation assays. Immunohistochemical staining in human PCa tissue microarrays revealed ATM expression is highly correlated with TR4 expression. Together, these results suggest TR4 may function as a tumor suppressor to prevent or delay prostate tumorigenesis via regulating ATM expression at the transcriptional level.
Collapse
|
67
|
Prostatic inflammation enhances basal-to-luminal differentiation and accelerates initiation of prostate cancer with a basal cell origin. Proc Natl Acad Sci U S A 2013; 111:E592-600. [PMID: 24367088 DOI: 10.1073/pnas.1318157111] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chronic inflammation has been shown to promote the initiation and progression of diverse malignancies by inducing genetic and epigenetic alterations. In this study, we investigate an alternative mechanism through which inflammation promotes the initiation of prostate cancer. Adult murine prostate epithelia are composed predominantly of basal and luminal cells. Previous studies revealed that the two lineages are largely self-sustained when residing in their native microenvironment. To interrogate whether tissue inflammation alters the differentiation program of basal cells, we conducted lineage tracing of basal cells using a K14-CreER;mTmG model in concert with a murine model of prostatitis induced by infection from the uropathogenic bacteria CP9. We show that acute prostatitis causes tissue damage and creates a tissue microenvironment that induces the differentiation of basal cells into luminal cells, an alteration that rarely occurs under normal physiological conditions. Previously we showed that a mouse model with prostate basal cell-specific deletion of Phosphatase and tensin homolog (K14-CreER;Pten(fl/fl)) develops prostate cancer with a long latency, because disease initiation in this model requires and is limited by the differentiation of transformation-resistant basal cells into transformation-competent luminal cells. Here, we show that CP9-induced prostatitis significantly accelerates the initiation of prostatic intraepithelial neoplasia in this model. Our results demonstrate that inflammation results in a tissue microenvironment that alters the normal prostate epithelial cell differentiation program and that through this cellular process inflammation accelerates the initiation of prostate cancer with a basal cell origin.
Collapse
|
68
|
Zhao H, Bauzon F, Fu H, Lu Z, Cui J, Nakayama K, Nakayama KI, Locker J, Zhu L. Skp2 deletion unmasks a p27 safeguard that blocks tumorigenesis in the absence of pRb and p53 tumor suppressors. Cancer Cell 2013; 24:645-59. [PMID: 24229711 PMCID: PMC3880806 DOI: 10.1016/j.ccr.2013.09.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/22/2013] [Accepted: 09/30/2013] [Indexed: 01/07/2023]
Abstract
pRb and p53 are two major tumor suppressors. Here, we found that p53 activates expression of Pirh2 and KPC1, two of the three ubiquitin ligases for p27. Loss of p53 in the absence of Skp2, the third ubiquitin ligase for p27, shrinks the cellular pool of p27 ubiquitin ligases to accumulate p27 protein. In the absence of pRb and p53, p27 was unable to inhibit DNA synthesis in spite of its abundance, but could inhibit division of cells that maintain DNA replication with rereplication. This mechanism blocked pRb/p53 doubly deficient pituitary and prostate tumorigenesis lastingly coexistent with bromodeoxyuridine-labeling neoplastic lesions, revealing an unconventional cancer cell vulnerability when pRb and p53 are inactivated.
Collapse
Affiliation(s)
- Hongling Zhao
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Frederick Bauzon
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hao Fu
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zhonglei Lu
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jinhua Cui
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Keiko Nakayama
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Keiich I. Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Liang Zhu
- Department of Developmental and Molecular Biology, and Medicine, and Pathology2, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
69
|
ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer. Proc Natl Acad Sci U S A 2013; 110:E3506-15. [PMID: 23918374 DOI: 10.1073/pnas.1303558110] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Combinatorial activation of PI3-kinase and RAS signaling occurs frequently in advanced prostate cancer and is associated with adverse patient outcome. We now report that the oncogenic Ets variant 4 (Etv4) promotes prostate cancer metastasis in response to coactivation of PI3-kinase and Ras signaling pathways in a genetically engineered mouse model of highly penetrant, metastatic prostate cancer. Using an inducible Cre driver to simultaneously inactivate Pten while activating oncogenic Kras and a fluorescent reporter allele in the prostate epithelium, we performed lineage tracing in vivo to define the temporal and spatial occurrence of prostate tumors, disseminated tumor cells, and metastases. These analyses revealed that though disseminated tumors cells arise early following the initial occurrence of prostate tumors, there is a significant temporal lag in metastasis, which is temporally coincident with the up-regulation of Etv4 expression in primary tumors. Functional studies showed that knockdown of Etv4 in a metastatic cell line derived from the mouse model abrogates the metastatic phenotype but does not affect tumor growth. Notably, expression and activation of ETV4, but not other oncogenic ETS genes, is correlated with activation of both PI3-kinase and Ras signaling in human prostate tumors and metastases. Our findings indicate that ETV4 promotes metastasis in prostate tumors that have activation of PI3-kinase and Ras signaling, and therefore, ETV4 represents a potential target of therapeutic intervention for metastatic prostate cancer.
Collapse
|
70
|
Loss of survivin in the prostate epithelium impedes carcinogenesis in a mouse model of prostate adenocarcinoma. PLoS One 2013; 8:e69484. [PMID: 23936028 PMCID: PMC3729965 DOI: 10.1371/journal.pone.0069484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/10/2013] [Indexed: 11/19/2022] Open
Abstract
The inhibitor of apoptosis protein survivin is expressed in most cancers. Using the conditional PTEN deletion mouse model, we previously reported that survivin levels increase with prostate tumor growth. Here we evaluated the functional role of survivin in prostate tumor growth. First, we demonstrated that mice lacking the survivin gene in prostate epithelium were fertile and had normal prostate growth and development. We then serially, from about 10-56 weeks of age, evaluated histopathologic changes in the prostate of mice with PTEN deletion combined with survivin mono- or bi-allelic gene deletion. While within this time period most of the animals with wild-type or monoallelic survivin deletion developed adenocarcinomas, the most severe lesions in the biallelic survivin deleted mice were high-grade prostatic intra-epithelial neoplasia with distinct histopathology. Many atypical cells contained large hypertrophic cytoplasm and desmoplastic reaction in the prostatic intra-epithelial neoplasia lesions of this group was minimal until the late ages. A reduced proliferation index as well as apoptotic and senescent cells were detected in the lesions of mice with compound PTEN/survivin deficiency throughout the time points examined. Survivin deletion was also associated with reduced tumor expression of another inhibitor of apoptosis member, the X-linked inhibitor of apoptosis. Our findings suggest that survivin participates in the progression of prostatic intraepithelial neoplasia to adenocarcinoma, and that survivin interference at the prostatic intraepithelial neoplasia stages may be a potential therapeutic strategy to halt or delay further progression.
Collapse
|
71
|
Zhang B, Chen H, Zhang L, Dakhova O, Zhang Y, Lewis MT, Creighton CJ, Ittmann MM, Xin L. A dosage-dependent pleiotropic role of Dicer in prostate cancer growth and metastasis. Oncogene 2013; 33:3099-108. [PMID: 23851498 PMCID: PMC3916938 DOI: 10.1038/onc.2013.281] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/25/2013] [Accepted: 05/24/2013] [Indexed: 12/12/2022]
Abstract
Dicer is as an RNase III enzyme essential for the maturation of the majority of microRNAs. Recent studies have revealed down-regulation or hemizygous loss of Dicer in many tumor models and demonstrated that suppressing Dicer activity enhances tumorigenic activities of lung and breast cancer cells, which support Dicer as a haploinsufficient tumor suppressor in these cancer models. Surprisingly, we found that knocking down Dicer expression suppresses the growth and tumorigenic capacity of human prostate cancer cell lines, but enhances migratory capacities of some prostate cancer cell lines. Dicer is up-regulated in human prostate cancer specimens, but lower Dicer expression portends a shorter time to recurrence. Complete ablation of Dicer activity in a Pten null mouse model for prostate cancer significantly halts tumor growth and progression, demonstrating that microRNAs play a critical role in maintaining cancer cell fitness. In comparison, hemizygous loss of Dicer in the same model also reduces primary tumor burden, but induces a more locally invasive phenotype and causes seminal vesicle obstruction at high penetrance. Disrupting Dicer activity leads to an increase in apoptosis and senescence in these models, presumably through up-regulation of P16/INK4a and P27/Kip1. Collectively, these results highlight a pleotropic role of Dicer in tumorigenesis that is not only dosage-dependent but also tissue context-dependent.
Collapse
Affiliation(s)
- B Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - H Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - L Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - O Dakhova
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Y Zhang
- Dan L. Duncan Cancer Center, Houston, TX, USA
| | - M T Lewis
- 1] Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA [2] Dan L. Duncan Cancer Center, Houston, TX, USA
| | | | - M M Ittmann
- 1] Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [2] Dan L. Duncan Cancer Center, Houston, TX, USA
| | - L Xin
- 1] Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA [2] Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [3] Dan L. Duncan Cancer Center, Houston, TX, USA
| |
Collapse
|
72
|
Xing C, Fu X, Sun X, Guo P, Li M, Dong JT. Different expression patterns and functions of acetylated and unacetylated Klf5 in the proliferation and differentiation of prostatic epithelial cells. PLoS One 2013; 8:e65538. [PMID: 23755247 PMCID: PMC3673967 DOI: 10.1371/journal.pone.0065538] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/25/2013] [Indexed: 12/18/2022] Open
Abstract
KLF5 is a basic transcription factor that regulates multiple biological processes. While it was identified as a putative tumor suppressor in prostate cancer, likely due to its function as an effector of TGF-β in the inhibition of cell proliferation, KLF5 is unacetylated and promotes cell proliferation in the absence of TGF-β. In this study, we evaluated the expression and function of KLF5 in prostatic epithelial homeostasis and tumorigenesis using mouse prostates and human prostate epithelial cells in 3-D culture. Histological and molecular analyses demonstrated that unacetylated-Klf5 was expressed in basal or undifferentiated cells, whereas acetylated-Klf5 was expressed primarily in luminal and/or differentiated cells. Androgen depletion via castration increased both the level of Klf5 expression and the number of Klf5-positive cells in the remaining prostate. Functionally, knockdown of KLF5 in the human RWPE-1 prostate cell line decreased the number of spheres formed in 3-D culture. In addition, knockout of Klf5 in prostate epithelial cells, mediated by probasin promoter-driven Cre expression, did not cause neoplasia but promoted cell proliferation and induced hyperplasia when one Klf5 allele was knocked out. Knockout of both Klf5 alleles however, caused apoptosis rather than cell proliferation in the epithelium. In castrated mice, knockout of Klf5 resulted in more severe shrinkage of the prostate. These results suggest that KLF5 plays a role in the proliferation and differentiation of prostatic epithelial cells, yet loss of KLF5 alone is insufficient to induce malignant transformation in epithelial cells.
Collapse
Affiliation(s)
- Changsheng Xing
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Peng Guo
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mei Li
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
73
|
Irshad S, Abate-Shen C. Modeling prostate cancer in mice: something old, something new, something premalignant, something metastatic. Cancer Metastasis Rev 2013; 32:109-22. [PMID: 23114843 PMCID: PMC3584242 DOI: 10.1007/s10555-012-9409-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
More than 15 years ago, the first generation of genetically engineered mouse (GEM) models of prostate cancer was introduced. These transgenic models utilized prostate-specific promoters to express SV40 oncogenes specifically in prostate epithelium. Since the description of these initial models, there have been a plethora of GEM models of prostate cancer representing various perturbations of oncogenes or tumor suppressors, either alone or in combination. This review describes these GEM models, focusing on their relevance for human prostate cancer and highlighting their strengths and limitations, as well as opportunities for the future.
Collapse
Affiliation(s)
- Shazia Irshad
- Herbert Irving Comprehensive Cancer Center, Departments of Urology and Pathology & Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | |
Collapse
|
74
|
Pommier AJC, Dufour J, Alves G, Viennois E, De Boussac H, Trousson A, Volle DH, Caira F, Val P, Arnaud P, Lobaccaro JMA, Baron S. Liver x receptors protect from development of prostatic intra-epithelial neoplasia in mice. PLoS Genet 2013; 9:e1003483. [PMID: 23675307 PMCID: PMC3649972 DOI: 10.1371/journal.pgen.1003483] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 03/14/2013] [Indexed: 12/13/2022] Open
Abstract
LXR (Liver X Receptors) act as “sensor” proteins that regulate cholesterol uptake, storage, and efflux. LXR signaling is known to influence proliferation of different cell types including human prostatic carcinoma (PCa) cell lines. This study shows that deletion of LXR in mouse fed a high-cholesterol diet recapitulates initial steps of PCa development. Elevation of circulating cholesterol in Lxrαβ-/- double knockout mice results in aberrant cholesterol ester accumulation and prostatic intra-epithelial neoplasia. This phenotype is linked to increased expression of the histone methyl transferase EZH2 (Enhancer of Zeste Homolog 2), which results in the down-regulation of the tumor suppressors Msmb and Nkx3.1 through increased methylation of lysine 27 of histone H3 (H3K27) on their promoter regions. Altogether, our data provide a novel link between LXR, cholesterol homeostasis, and epigenetic control of tumor suppressor gene expression. Cholesterol is one of the major metabolic molecules required for a broad range of cellular processes. Recent advances in prostate cancer research have demonstrated that tumor cells need to increase their supply of cholesterol to sustain membrane building, proliferation, and survival capacities. Liver X receptors, which belong to the nuclear receptor superfamily, are central mediators of cholesterol homeostasis. Indeed, they regulate the expression of many genes involved in cholesterol uptake storage and efflux. Here, we show that genetic ablation of LXRs in mice results in the formation of precancerous lesions in the prostate, called prostatic intra-epithelial neoplasia. These are only observed when mice are fed a high-cholesterol diet. Hence, LXRs regulate cholesterol homeostasis in the prostate and protect cells from abnormal proliferation when exposed to high dietary cholesterol.
Collapse
Affiliation(s)
- Aurélien J. C. Pommier
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Julie Dufour
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Georges Alves
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Emilie Viennois
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Hugues De Boussac
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Amalia Trousson
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - David H. Volle
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Françoise Caira
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Pierre Val
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
| | - Philippe Arnaud
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
| | - Jean-Marc A. Lobaccaro
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Silvère Baron
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, BP 10448, Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
- * E-mail:
| |
Collapse
|
75
|
Wu TY, Khor TO, Su ZY, Saw CLL, Shu L, Cheung KL, Huang Y, Yu S, Kong ANT. Epigenetic modifications of Nrf2 by 3,3'-diindolylmethane in vitro in TRAMP C1 cell line and in vivo TRAMP prostate tumors. AAPS JOURNAL 2013; 15:864-74. [PMID: 23658110 DOI: 10.1208/s12248-013-9493-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 04/17/2013] [Indexed: 01/09/2023]
Abstract
3,3'-diindolylmethane (DIM) is currently being investigated in many clinical trials including prostate, breast, and cervical cancers and has been shown to possess anticancer effects in several in vivo and in vitro models. Previously, DIM has been reported to possess cancer chemopreventive effects in prostate carcinogenesis in TRAMP mice; however, the in vivo mechanism is unclear. The present study aims to investigate the in vitro and in vivo epigenetics modulation of DIM in TRAMP-C1 cells and in TRAMP mouse model. In vitro study utilizing TRAMP-C1 cells showed that DIM suppressed DNMT expression and reversed CpG methylation status of Nrf2 resulting in enhanced expression of Nrf2 and Nrf2-target gene NQO1. In vivo study, TRAMP mice fed with DIM-supplemented diet showed much lower incidence of tumorigenesis and metastasis than the untreated control group similar to what was reported previously. DIM increased apoptosis, decreased cell proliferation and enhanced Nrf2 and Nrf2-target gene NQO1 expression in prostate tissues. Importantly, immunohistochemical analysis showed that DIM reduced the global CpG 5-methylcytosine methylation. Focusing on one of the early cancer chemopreventive target gene Nrf2, bisulfite genomic sequencing showed that DIM decreased the methylation status of the first five CpGs of the Nrf2 promoter region, corroborating with the results of in vitro TRAMP-C1 cells. In summary, our current study shows that DIM is a potent cancer chemopreventive agent for prostate cancer and epigenetic modifications of the CpG including Nrf2 could be a potential mechanism by which DIM exerts its chemopreventive effects.
Collapse
Affiliation(s)
- Tien-Yuan Wu
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Ittmann M, Huang J, Radaelli E, Martin P, Signoretti S, Sullivan R, Simons BW, Ward JM, Robinson BD, Chu GC, Loda M, Thomas G, Borowsky A, Cardiff RD. Animal models of human prostate cancer: the consensus report of the New York meeting of the Mouse Models of Human Cancers Consortium Prostate Pathology Committee. Cancer Res 2013; 73:2718-36. [PMID: 23610450 DOI: 10.1158/0008-5472.can-12-4213] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Animal models, particularly mouse models, play a central role in the study of the etiology, prevention, and treatment of human prostate cancer. While tissue culture models are extremely useful in understanding the biology of prostate cancer, they cannot recapitulate the complex cellular interactions within the tumor microenvironment that play a key role in cancer initiation and progression. The National Cancer Institute (NCI) Mouse Models of Human Cancers Consortium convened a group of human and veterinary pathologists to review the current animal models of prostate cancer and make recommendations about the pathologic analysis of these models. More than 40 different models with 439 samples were reviewed, including genetically engineered mouse models, xenograft, rat, and canine models. Numerous relevant models have been developed over the past 15 years, and each approach has strengths and weaknesses. Analysis of multiple genetically engineered models has shown that reactive stroma formation is present in all the models developing invasive carcinomas. In addition, numerous models with multiple genetic alterations display aggressive phenotypes characterized by sarcomatoid carcinomas and metastases, which is presumably a histologic manifestation of epithelial-mesenchymal transition. The significant progress in development of improved models of prostate cancer has already accelerated our understanding of the complex biology of prostate cancer and promises to enhance development of new approaches to prevention, detection, and treatment of this common malignancy.
Collapse
Affiliation(s)
- Michael Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Narlik-Grassow M, Blanco-Aparicio C, Cecilia Y, Perez M, Muñoz-Galvan S, Cañamero M, Carnero A. Conditional transgenic expression of PIM1 kinase in prostate induces inflammation-dependent neoplasia. PLoS One 2013; 8:e60277. [PMID: 23565217 PMCID: PMC3614961 DOI: 10.1371/journal.pone.0060277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/24/2013] [Indexed: 11/19/2022] Open
Abstract
The Pim proteins are a family of highly homologous protein serine/threonine kinases that have been found to be overexpressed in cancer. Elevated levels of Pim1 kinase were first discovered in human leukemia and lymphomas. However, more recently Pim1 was found to be increased in solid tumors, including pancreatic and prostate cancers, and has been proposed as a prognostic marker. Although the Pim kinases have been identified as oncogenes in transgenic models, they have weak transforming abilities on their own. However, they have been shown to greatly enhance the ability of other genes or chemical carcinogens to induce tumors. To explore the role of Pim1 in prostate cancer, we generated conditional Pim1 transgenic mice, expressed Pim1 in prostate epithelium, and analyzed the contribution of PIM1 to neoplastic initiation and progression. Accordingly, we explored the effect of PIM1 overexpression in 3 different settings: upon hormone treatment, during aging, and in combination with the absence of one Pten allele. We have found that Pim1 overexpression increased the severity of mouse prostate intraepithelial neoplasias (mPIN) moderately in all three settings. Furthermore, Pim1 overexpression, in combination with the hormone treatment, increased inflammation surrounding target tissues leading to pyelonephritis in transgenic animals. Analysis of senescence induced in these prostatic lesions showed that the lesions induced in the presence of inflammation exhibited different behavior than those induced in the absence of inflammation. While high grade prostate preneoplastic lesions, mPIN grades III and IV, in the presence of inflammation did not show any senescence markers and demonstrated high levels of Ki67 staining, untreated animals without inflammation showed senescence markers and had low levels of Ki67 staining in similar high grade lesions. Our data suggest that Pim1 might contribute to progression rather than initiation in prostate neoplasia.
Collapse
Affiliation(s)
- Maja Narlik-Grassow
- Experimental Therapeutics programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Yolanda Cecilia
- Experimental Therapeutics programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marco Perez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Cientificas, Universidad de Sevilla, Sevilla, Spain
| | - Sandra Muñoz-Galvan
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Cientificas, Universidad de Sevilla, Sevilla, Spain
| | - Marta Cañamero
- Biotechnology programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Cientificas, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
78
|
Effects of dietary high fat on prostate intraepithelial neoplasia in TRAMP mice. Lab Anim Res 2013; 29:39-47. [PMID: 23573107 PMCID: PMC3616208 DOI: 10.5625/lar.2013.29.1.39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/04/2013] [Accepted: 03/05/2013] [Indexed: 12/22/2022] Open
Abstract
Increased fat intake is known to be a major cause of prostate cancer. In this study, we investigated the effect of dietary high fat on prostate intraepithelial neoplasia using transgenic adenocarcinoma mouse prostate (TRAMP) mice. Six-week-old male TRAMP mice were fed AIN93G (control group, 4.0 kcal/kg, n=6) and AIN93G-HFD (experimental group, 4.8 kcal/kg, n=7) for 10 weeks. Prostate histopathology, urogenital tract (UGT) weight, epididymal white adipose tissue weight, argyrophilic nucleolar organizer regions (AgNORs) counts, and serum leptin levels were examined. AIN93G-HFD fed group showed progressed neoplastic lesions in the prostate (P<0.05) compared to AIN93G fed group. AIN93G-HFD intake resulted in a increase in the weight of UGT (P<0.05) and epididymal white adipose tissue. The number of Ag-NOR positive dots significantly increased in each prostate lobe and final serum leptin levels in AIN93G-HFD fed group were about twice those of AIN93G fed group (P<0.05). Dietary high fat was related to the prostate cancer progression in the early stage of TRAMP mice and increased serum leptin levels, suggesting that the regulation of dietary components could delay the progression of prostate cancer.
Collapse
|
79
|
Lineage analysis of basal epithelial cells reveals their unexpected plasticity and supports a cell-of-origin model for prostate cancer heterogeneity. Nat Cell Biol 2013; 15:274-83. [PMID: 23434823 PMCID: PMC3743266 DOI: 10.1038/ncb2697] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 01/21/2013] [Indexed: 12/24/2022]
Abstract
A key issue in cancer biology is whether oncogenic transformation of different cell types of origin within an adult tissue gives rise to distinct tumour subtypes that differ in their prognosis and/or treatment response. We now show that initiation of prostate tumours in basal or luminal epithelial cells in mouse models results in tumours with distinct molecular signatures that are predictive of human patient outcomes. Furthermore, our analysis of untransformed basal cells reveals an unexpected assay dependence of their stem cell properties in sphere formation and transplantation assays versus genetic lineage tracing during prostate regeneration and adult tissue homeostasis. Although oncogenic transformation of basal cells gives rise to tumours with luminal phenotypes, cross-species bioinformatic analyses indicate that tumours of luminal origin are more aggressive than tumours of basal origin, and identify a molecular signature associated with patient outcome. Our results reveal the inherent plasticity of basal cells, and support a model in which different cells of origin generate distinct molecular subtypes of prostate cancer.
Collapse
|
80
|
β-catenin is required for prostate development and cooperates with Pten loss to drive invasive carcinoma. PLoS Genet 2013; 9:e1003180. [PMID: 23300485 PMCID: PMC3536663 DOI: 10.1371/journal.pgen.1003180] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/04/2012] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is a major cause of male death in the Western world, but few frequent genetic alterations that drive prostate cancer initiation and progression have been identified. β-Catenin is essential for many developmental processes and has been implicated in tumorigenesis in many tissues, including prostate cancer. However, expression studies on human prostate cancer samples are unclear on the role this protein plays in this disease. We have used in vivo genetic studies in the embryo and adult to extend our understanding of the role of β-Catenin in the normal and neoplastic prostate. Our gene deletion analysis revealed that prostate epithelial β-Catenin is required for embryonic prostate growth and branching but is dispensable in the normal adult organ. During development, β-Catenin controls the number of progenitors in the epithelial buds and regulates a discrete network of genes, including c-Myc and Nkx3.1. Deletion of β-Catenin in a Pten deleted model of castration-resistant prostate cancer demonstrated it is dispensable for disease progression in this setting. Complementary overexpression experiments, through in vivo protein stabilization, showed that β-Catenin promotes the formation of squamous epithelia during prostate development, even in the absence of androgens. β-Catenin overexpression in combination with Pten loss was able to drive progression to invasive carcinoma together with squamous metaplasia. These studies demonstrate that β-Catenin is essential for prostate development and that an inherent property of high levels of this protein in prostate epithelia is to drive squamous fate differentiation. In addition, they show that β-Catenin overexpression can promote invasive prostate cancer in a clinically relevant model of this disease. These data provide novel information on cancer progression pathways that give rise to lethal prostate disease in humans. Prostate cancer is a major cause of male death in the Western world, but few genes involved in this disease have been identified. We have undertaken an in-depth in vivo analysis in the prostate of the β-Catenin protein, which has been shown to be important in many processes during embryogenesis and has been implicated in tumorigenesis. Our studies demonstrate that β-Catenin is essential for prostate development but is dispensable in the normal adult organ. Analysis of a mouse model of a frequently mutated human prostate tumour suppressor, Pten loss, revealed that β-Catenin is not required for neoplastic formation in this model, even in castrated conditions. However, increased β-Catenin levels can cooperate with Pten loss to promote the progression of aggressive invasive prostate cancer together with squamous metaplasia. These data uncover the role of β-Catenin in the prostate and provide new insights on how pathways interact to drive human prostate cancer.
Collapse
|
81
|
Creasy D, Bube A, de Rijk E, Kandori H, Kuwahara M, Masson R, Nolte T, Reams R, Regan K, Rehm S, Rogerson P, Whitney K. Proliferative and nonproliferative lesions of the rat and mouse male reproductive system. Toxicol Pathol 2013; 40:40S-121S. [PMID: 22949412 DOI: 10.1177/0192623312454337] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature and differential diagnosis for classifying microscopic lesions observed in the male reproductive system of laboratory rats and mice, with color microphotographs illustrating examples of some lesions. The standardized nomenclature presented in this document is also available for society members electronically on the Internet (http://goreni.org). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous and aging lesions as well as lesions induced by exposure to test materials. A widely accepted and utilized international harmonization of nomenclature for lesions of the male reproductive system in laboratory animals will decrease confusion among regulatory and scientific research organizations in different countries and provide a common language to increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
- Dianne Creasy
- Huntingdon Life Sciences, East Millstone, New Jersey 08875, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Li G, Rivas P, Bedolla R, Thapa D, Reddick RL, Ghosh R, Kumar AP. Dietary resveratrol prevents development of high-grade prostatic intraepithelial neoplastic lesions: involvement of SIRT1/S6K axis. Cancer Prev Res (Phila) 2012; 6:27-39. [PMID: 23248098 DOI: 10.1158/1940-6207.capr-12-0349] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIRT1 (mammalian ortholog of the yeast silent information regulator 2) is a NAD-dependent histone deacetylase belonging to the multigene family of sirtuins. Anecdotal and epidemiologic observations provide evidence for beneficial effects of the calorie restriction mimetic resveratrol (RES), a SIRT1 activator in preventing cardiovascular diseases and cancer. Although SIRT1 possesses both tumorigenic and antitumorigenic potential, the molecular mechanisms underlying SIRT1-mediated tumor progression or inhibition are poorly understood. In this study, we investigated the role of SIRT1 in multiple human prostate cancer cell lines and prostate-specific PTEN knockout mouse model using resveratrol. Androgen-independent prostate cancer cell lines (C42B, PC3, and DU145) express higher levels of SIRT1 than androgen-responsive (LNCaP) and nontumorigenic prostate cells (RWPE-1). Resveratrol enhanced this expression without any significant effect on SIRT1 enzymatic activity. Inhibition of SIRT1 expression using shRNA enhanced cell proliferation and inhibited autophagy by repressing phosphorylation of S6K and 4E-BP1. These biologic correlates were reversed in the presence of resveratrol. Analysis of prostates from dietary intervention with resveratrol showed a significant reduction in prostate weight and reduction in the incidence of high-grade prostatic intraepithelial neoplastic (HGPIN) lesions by approximately 54% with no significant change in body weight. Consistent with the in vitro findings, resveratrol intervention in the PTEN knockout mouse model was associated with reduction in the prostatic levels of mTOR complex 1 (mTORC1) activity and increased expression of SIRT1. These data suggest that SIRT1/S6K-mediated inhibition of autophagy drives prostate tumorigenesis. Therefore, modulation of SIRT1/S6K signaling represents an effective strategy for prostate cancer prevention.
Collapse
Affiliation(s)
- Guiming Li
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Qin J, Wu SP, Creighton CJ, Dai F, Xie X, Cheng CM, Frolov A, Ayala G, Lin X, Feng XH, Ittmann MM, Tsai SJ, Tsai MJ, Tsai SY. COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis. Nature 2012. [PMID: 23201680 DOI: 10.1038/nature11674] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mutations in phosphatase and tensin homologue (PTEN) or genomic alterations in the phosphatidylinositol-3-OH kinase-signalling pathway are the most common genetic alterations reported in human prostate cancer. However, the precise mechanism underlying how indolent tumours with PTEN alterations acquire metastatic potential remains poorly understood. Recent studies suggest that upregulation of transforming growth factor (TGF)-β signalling triggered by PTEN loss will form a growth barrier as a defence mechanism to constrain prostate cancer progression, underscoring that TGF-β signalling might represent a pre-invasive checkpoint to prevent PTEN-mediated prostate tumorigenesis. Here we show that COUP transcription factor II (COUP-TFII, also known as NR2F2), a member of the nuclear receptor superfamily, serves as a key regulator to inhibit SMAD4-dependent transcription, and consequently overrides the TGF-β-dependent checkpoint for PTEN-null indolent tumours. Overexpression of COUP-TFII in the mouse prostate epithelium cooperates with PTEN deletion to augment malignant progression and produce an aggressive metastasis-prone tumour. The functional counteraction between COUP-TFII and SMAD4 is reinforced by genetically engineered mouse models in which conditional loss of SMAD4 diminishes the inhibitory effects elicited by COUP-TFII ablation. The biological significance of COUP-TFII in prostate carcinogenesis is substantiated by patient sample analysis, in which COUP-TFII expression or activity is tightly correlated with tumour recurrence and disease progression, whereas it is inversely associated with TGF-β signalling. These findings reveal that the destruction of the TGF-β-dependent barrier by COUP-TFII is crucial for the progression of PTEN-mutant prostate cancer into a life-threatening disease, and supports COUP-TFII as a potential drug target for the intervention of metastatic human prostate cancer.
Collapse
Affiliation(s)
- Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Floc'h N, Kinkade CW, Kobayashi T, Aytes A, Lefebvre C, Mitrofanova A, Cardiff RD, Califano A, Shen MM, Abate-Shen C. Dual targeting of the Akt/mTOR signaling pathway inhibits castration-resistant prostate cancer in a genetically engineered mouse model. Cancer Res 2012; 72:4483-93. [PMID: 22815528 PMCID: PMC3432676 DOI: 10.1158/0008-5472.can-12-0283] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the prognosis for clinically localized prostate cancer is now favorable, there are still no curative treatments for castration-resistant prostate cancer (CRPC) and, therefore, it remains fatal. In this study, we investigate a new therapeutic approach for treatment of CRPC, which involves dual targeting of a major signaling pathway that is frequently deregulated in the disease. We found that dual targeting of the Akt and mTOR signaling pathways with their respective inhibitors, MK-2206 and ridaforolimus (MK-8669), is highly effective for inhibiting CRPC in preclinical studies in vivo using a refined genetically engineered mouse model of the disease. The efficacy of the combination treatment contrasts with their limited efficacy as single agents, since delivery of MK-2206 or MK-8669 individually had a modest impact in vivo on the overall tumor phenotype. In human prostate cancer cell lines, although not in the mouse model, the synergistic actions of MK-2206 and ridaforolimus (MK-8669) are due in part to limiting the mTORC2 feedback activation of Akt. Moreover, the effects of these drugs are mediated by inhibition of cellular proliferation via the retinoblastoma (Rb) pathway. Our findings suggest that dual targeting of the Akt and mTOR signaling pathways using MK-2206 and ridaforolimus (MK-8669) may be effective for treatment of CRPC, particularly for patients with deregulated Rb pathway activity.
Collapse
Affiliation(s)
- Nicolas Floc'h
- Departments of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Carolyn Waugh Kinkade
- Departments of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Takashi Kobayashi
- Departments of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Alvaro Aytes
- Departments of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Celine Lefebvre
- Department of Biomedical Informatics and Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Antonina Mitrofanova
- Department of Biomedical Informatics and Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Robert D. Cardiff
- Center for Comparative Medicine and Department of Pathology, School of Medicine, University of California, Davis 95616
| | - Andrea Califano
- Department of Biomedical Informatics and Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Michael M. Shen
- Departments of Medicine and Genetics & Development, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| | - Cory Abate-Shen
- Departments of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
85
|
Wang J, Kobayashi T, Floc'h N, Kinkade CW, Aytes A, Dankort D, Lefebvre C, Mitrofanova A, Cardiff RD, McMahon M, Califano A, Shen MM, Abate-Shen C. B-Raf activation cooperates with PTEN loss to drive c-Myc expression in advanced prostate cancer. Cancer Res 2012; 72:4765-76. [PMID: 22836754 DOI: 10.1158/0008-5472.can-12-0820] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Both the PI3K → Akt → mTOR and mitogen-activated protein kinase (MAPK) signaling pathways are often deregulated in prostate tumors with poor prognosis. Here we describe a new genetically engineered mouse model of prostate cancer in which PI3K-Akt-mTOR signaling is activated by inducible disruption of PTEN, and extracellular signal-regulated kinase 1/2 (ERK1/2) MAPK signaling is activated by inducible expression of a BRAF(V600E) oncogene. These tissue-specific compound mutant mice develop lethal prostate tumors that are inherently resistant to castration. These tumors bypass cellular senescence and disseminate to lymph nodes, bone marrow, and lungs where they form overt metastases in approximately 30% of the cases. Activation of PI3K → Akt → mTOR and MAPK signaling pathways in these prostate tumors cooperate to upregulate c-Myc. Accordingly, therapeutic treatments with rapamycin and PD0325901 to target these pathways, respectively, attenuate c-Myc levels and reduce tumor and metastatic burden. Together, our findings suggest a generalized therapeutic approach to target c-Myc activation in prostate cancer by combinatorial targeting of the PI3K → Akt → mTOR and ERK1/2 MAPK signaling pathways.
Collapse
Affiliation(s)
- Jingqiang Wang
- Department of Urology and Pathology and Cell Biology, Columbia University Medical Center, New York, New York 10031, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Lai KP, Yamashita S, Huang CK, Yeh S, Chang C. Loss of stromal androgen receptor leads to suppressed prostate tumourigenesis via modulation of pro-inflammatory cytokines/chemokines. EMBO Mol Med 2012; 4:791-807. [PMID: 22745041 PMCID: PMC3494077 DOI: 10.1002/emmm.201101140] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 03/05/2012] [Accepted: 04/05/2012] [Indexed: 12/12/2022] Open
Abstract
Stromal-epithelial interaction is crucial to mediate normal prostate and prostate cancer (PCa) development. The indispensable roles of mesenchymal/stromal androgen receptor (AR) for the prostate organogenesis have been demonstrated by using tissue recombination from wild-type and testicular feminized mice. However, the stromal AR functions in the tumour microenvironment and the underlying mechanisms governing the interactions between the epithelium and stroma are not completely understood. Here, we have established the first animal model with AR deletion in stromal fibromuscular cells (dARKO, AR knockout in fibroblasts and smooth muscle cells) in the Pten(+/-) mouse model that can spontaneously develop prostatic intraepithelial neoplasia (PIN). We found that loss of stromal fibromuscular AR led to suppression of PIN lesion development with alleviation of epithelium proliferation and tumour-promoting microenvironments, including extracellular matrix (ECM) remodelling, immune cell infiltration and neovasculature formation due, in part, to the modulation of pro-inflammatory cytokines/chemokines. Finally, targeting stromal fibromuscular AR with the AR degradation enhancer, ASC-J9®, resulted in the reduction of PIN development/progression, which might provide a new approach to suppress PIN development.
Collapse
Affiliation(s)
- Kuo-Pao Lai
- Departments of Pathology, Urology, and Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | |
Collapse
|
87
|
Li G, Wang H, Liu AB, Cheung C, Reuhl KR, Bosland MC, Yang CS. Dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine-induced prostate carcinogenesis in CYP1A-humanized mice. Cancer Prev Res (Phila) 2012; 5:963-72. [PMID: 22581815 DOI: 10.1158/1940-6207.capr-12-0023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To develop a relevant mouse model for prostate cancer prevention research, we administered a dietary carcinogen, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), to CYP1A-humanized mice. In comparison with mouse Cyp1a2, human CYP1A2 preferentially activates PhIP to a proximate carcinogen. Following a single oral dose of PhIP (200 mg/kg body weight), we observed inflammation, atrophy of acini, low-grade prostatic intraepithelial neoplasia (PIN; after 20 weeks), and high-grade PIN (HgPIN; after 30 to 50 weeks) in dorsolateral, ventral, and coagulating anterior prostate glands of these mice. These lesions were androgen receptor positive and featured the loss of expression of the basal cell marker p63 and the tumor suppressor PTEN. Similar to human prostate carcinogenesis, glutathione S-transferase P1 (GSTP1) expression was lost or partially lost in HgPIN. E-Cadherin expression was also lost in HgPIN. The expression of DNA methyltransferase 1 was elevated, possibly to enhance promoter hypermethylation for the silencing of GSTP1 and E-cadherin. Prostate carcinogenesis was promoted by a high-fat stress diet, resulting in HgPIN that developed earlier and in advanced lesions displayed features consistent with carcinoma in situ. This dietary carcinogen-induced prostate cancer model, recapitulating important features of early human prostate carcinogenesis, constitutes a new experimental system for prostate cancer research.
Collapse
Affiliation(s)
- Guangxun Li
- Department of Chemical Biology, Center for Cancer Prevention Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
88
|
Vinall RL, Chen JQ, Hubbard NE, Sulaimon SS, Shen MM, Devere White RW, Borowsky AD. Initiation of prostate cancer in mice by Tp53R270H: evidence for an alternative molecular progression. Dis Model Mech 2012; 5:914-20. [PMID: 22563073 PMCID: PMC3484872 DOI: 10.1242/dmm.008995] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Tp53 mutations are common in human prostate cancer (CaP), occurring with a frequency of ∼30% and ∼70% in localized and metastatic disease, respectively. In vitro studies have determined several common mutations of Tp53 that have specific gain-of-function properties in addition to loss of function, including the ability to promote castration-resistant (CR) growth of CaP cells in some contexts. To date, a lack of suitable mouse models has prohibited investigation of the role played by Tp53 mutations in mediating CaP progression in vivo. Here, we describe the effects of conditional expression of a mutant Tp53 (Tp53R270H; equivalent to the human hotspot mutant R273H) in the prostate epithelium of mice. Heterozygous “Tp53LSL-R270H/+” [129S4(Trp53tm3Tyj)] and “Nkx3.1-Cre” [129S(Nkx3-1tm3(cre)Mms)] mice with prostate-specific expression of the Tp53R270H mutation (p53R270H/+Nkx3.1-Cre mice) were bred onto an FVB/N background via speed congenesis to produce strain FVB.129S4(Trp53tm3Tyj/wt); FVB.129S(Nkx3-1tm3(cre)Mms/wt) and littermate genotype negative control mice. These mutant mice had significantly increased incidences of prostatic intraepithelial neoplasia (PIN) lesions, and these appeared earlier, compared with the Nkx3.1 haploinsufficient (Nkx3.1-Cre het) littermate mice, which did not express the Tp53 mutation. PIN lesions in these mice showed consistent progression and some developed into invasive adenocarcinoma with a high grade, sarcomatoid or epithelial-mesenchymal transition (EMT) phenotype. PIN lesions were similar to those seen in PTEN conditional knockout mice, with evidence of AKT activation concomitant with neoplastic proliferation. However, the invasive tumor phenotype is rarely seen in previously described mouse models of prostatic neoplasia. These data indicate that the Tp53R270H mutation plays a role in CaP initiation. This finding has not previously been reported. Further characterization of this model, particularly in a setting of androgen deprivation, should allow further insight into the mechanisms by which the Tp53R270H mutation mediates CaP progression.
Collapse
Affiliation(s)
- Ruth L Vinall
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Rancho Cordova, CA, USA
| | | | | | | | | | | | | |
Collapse
|
89
|
Hensley PJ, Kyprianou N. Modeling prostate cancer in mice: limitations and opportunities. JOURNAL OF ANDROLOGY 2012; 33:133-44. [PMID: 21680808 PMCID: PMC3726197 DOI: 10.2164/jandrol.111.013987] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The complex dynamics of the tumor microenvironment and prostate cancer heterogeneity have confounded efforts to establish suitable preclinical mouse models to represent human cancer progression from early proliferative phenotypes to aggressive, androgen-independent, and invasive metastatic tumors. Current models have been successful in capitulating individual characteristics of the aggressive tumors. However, none of these models comprehensively mimics human cancer progression, establishing the challenge in their exploitation to study human disease. The ability to tailor phenotypic outcomes in mice by compounding mutations to target specific molecular pathways provides a powerful tool toward disruption of signaling pathways contributing to the initiation and progression of castration-resistant prostate cancer. Each model is characterized by unique features contributing to the understanding of prostate tumorigenesis, as well as limitations challenging our knowledge of the mechanisms of cancer development and progression. Emerging strategies utilize genomic manipulation technology to circumvent these limitations toward the formulation of attractive, physiologically relevant models of prostate cancer progression to advanced disease. This review discusses the current value of the widely used and well-characterized mouse models of prostate cancer progression to metastasis, as well as the opportunities begging exploitation for the development of new models for testing the antitumor efficacy of therapeutic strategies and identifying new biomarkers of disease progression.
Collapse
Affiliation(s)
- Patrick J Hensley
- Department of Surgery/Urology, University of Kentucky, Lexington, KY 40536, USA
| | | |
Collapse
|
90
|
Choi N, Zhang B, Zhang L, Ittmann M, Xin L. Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell 2012; 21:253-65. [PMID: 22340597 PMCID: PMC3285423 DOI: 10.1016/j.ccr.2012.01.005] [Citation(s) in RCA: 253] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/02/2012] [Accepted: 01/06/2012] [Indexed: 12/21/2022]
Abstract
The prostate epithelial lineage hierarchy and the cellular origin for prostate cancer remain inadequately defined. Using a lineage-tracing approach, we show that adult rodent prostate basal and luminal cells are independently self-sustained in vivo. Disrupting the tumor suppressor Pten in either lineage led to prostate cancer initiation. However, the cellular composition and onset dynamics of the resulting tumors are distinctive. Prostate luminal cells are more responsive to Pten null-induced mitogenic signaling. In contrast, basal cells are resistant to direct transformation. Instead, loss of Pten activity induces the capability of basal cells to differentiate into transformation-competent luminal cells. Our study suggests that deregulation of epithelial differentiation is a critical step for the initiation of prostate cancers of basal cell origin.
Collapse
Affiliation(s)
- Nahyun Choi
- Department of Molecular and Cellular Biology
| | - Boyu Zhang
- Department of Molecular and Cellular Biology
| | - Li Zhang
- Department of Molecular and Cellular Biology
| | - Michael Ittmann
- Department of Pathology and Immunology
- Dan L. Duncan Cancer Center
| | - Li Xin
- Department of Molecular and Cellular Biology
- Department of Pathology and Immunology
- Dan L. Duncan Cancer Center
- Corresponding author: Li Xin, Ph.D., Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, Phone: 713-798-1650, FAX: 713-798-3017,
| |
Collapse
|
91
|
Wu TY, Saw CLL, Khor TO, Pung D, Boyanapalli SSS, Kong ANT. In vivo pharmacodynamics of indole-3-carbinol in the inhibition of prostate cancer in transgenic adenocarcinoma of mouse prostate (TRAMP) mice: involvement of Nrf2 and cell cycle/apoptosis signaling pathways. Mol Carcinog 2011; 51:761-70. [PMID: 21837756 DOI: 10.1002/mc.20841] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/11/2011] [Accepted: 07/13/2011] [Indexed: 12/23/2022]
Abstract
Indole-3-carbinol (I3C) found abundantly in crucifers has been shown to possess anti-cancer effects. The present study aims to examine the chemopreventive effects and the molecular mechanism of I3C, particularly the anti-oxidative stress pathway regulated by nuclear erythroid related factor 2 (Nrf2). HepG2-C8-ARE-luciferase cells were used for Nrf2-ARE activity. TRAMP C1 cells were used to investigate the effects of I3C on Nrf2-mediated genes. To test the chemopreventive efficacy of I3C, transgenic adenocarcinoma of mouse prostate (TRAMP) mice were fed with 1% I3C supplemented diet for 12 or 16 wk. The expression of Nrf2 and its downstream target genes, cell cycle and apoptosis genes were investigated using quantitative real-time polymerase chain reaction (qPCR). The protein expressions of these biomarkers were also investigated using Western blotting. I3C induced antioxidant response element (ARE)-luciferase activity in a dose-dependent manner. Treatments of TRAMP C1 cells with I3C also resulted in the induction of Nrf2-mediated genes. I3C significantly suppressed the incidence of palpable tumor and reduced the genitourinary weight in TRAMP mice. Western blots and qPCR analyses of prostate tissues showed that I3C induced the expression of Nrf2, NAD(P)H quinine oxidoreductase 1 (NQO-1) as well as cell cycle and apoptosis related biomarkers in I3C-fed TRAMP mice. This study demonstrated that the effectiveness of I3C as prostate cancer chemoprevention agent via up-regulation of a novel Nrf2-mediated anti-oxidative stress pathway.
Collapse
Affiliation(s)
- Tien-Yuan Wu
- Center for Cancer Prevention Research and Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
The serine/threonine kinase Akt is frequently activated in human cancers and is considered an attractive therapeutic target. However, the relative contributions of the different Akt isoforms to tumorigenesis, and the effect of their deficiencies on cancer development are not well understood. We had previously shown that Akt1 deficiency is sufficient to markedly reduce the incidence of tumors in Pten+/− mice. Particularly, Akt1 deficiency inhibits endometrial carcinoma and prostate neoplasia in Pten+/− mice. Here, we analyzed the effect of Akt2 deficiency on the incidence of tumors in Pten+/− mice. Relative to Akt1, Akt2 deficiency had little-to-no effect on the incidence of prostate neoplasia, endometrial carcinoma, intestinal polyps and adrenal lesions in Pten+/− mice. However, Akt2 deficiency significantly decreased the incidence of thyroid tumors in Pten+/−, which correlates with the relatively high level of Akt2 expression in the thyroid. Thus, unlike Akt1 deletion, Akt2 deletion is not sufficient to markedly inhibit tumorigenesis in Pten+/− mice in most tested tissues. The relatively small effect of Akt2 deletion on the inhibition of tumorigenesis in Pten+/− mice could be explained, in part, by an insufficient decrease in total Akt activity, due to the relatively lower Akt2 versus Akt1 expression, and relatively high blood insulin levels in Pten+/−Akt2−/− mice. The relatively high blood insulin levels in Pten+/−Akt2−/− mice may elevate the activity of Akt1, and possibly Akt3, thus, limiting the reduction of total Akt activity and preventing this activity from dropping to a threshold level required to inhibit tumorigenesis.
Collapse
|
93
|
Differentiation of the ductal epithelium and smooth muscle in the prostate gland are regulated by the Notch/PTEN-dependent mechanism. Dev Biol 2011; 356:337-49. [PMID: 21624358 DOI: 10.1016/j.ydbio.2011.05.659] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 05/11/2011] [Accepted: 05/12/2011] [Indexed: 02/08/2023]
Abstract
We have shown previously that during branching morphogenesis of the mouse prostate gland, Bone morphogenetic protein 7 functions to restrict Notch1-positive progenitor cells to the tips of the prostate buds. Here, we employed prostate-specific murine bi-genic systems to investigate the effects of gain and loss of Notch function during prostate development. We show that Nkx3.1(Cre) and Probasin(Cre) alleles drive expression of Cre recombinase to the prostate epithelium and periepithelial stroma. We investigated the effects of gain of Notch function using the Rosa(NI1C) conditional allele, which carries a constitutively active intracellular domain of Notch1 receptor. We carried out the analysis of loss of Notch function in Nkx3.1(Cre/+);RBP-J(flox/flox) prostates, where RBP-J is a ubiquitous transcriptional mediator of Notch signaling. We found that gain of Notch function resulted in inhibition of the tumor suppressor PTEN, and increase in cell proliferation and progenitor cells in the basal epithelium and smooth muscle compartments. In turn, loss of Notch/RBP-J function resulted in decreased cell proliferation and loss of epithelial and smooth muscle progenitors. Gain of Notch function resulted in an early onset of benign prostate hyperplasia by three months of age. Loss of Notch function also resulted in abnormal differentiation of the prostate epithelium and stroma. In particular, loss of Notch signaling and increase in PTEN promoted a switch from myoblast to fibroblast lineage, and a loss of smooth muscle. In summary, we show that Notch signaling is necessary for terminal differentiation of the prostate epithelium and smooth muscle, and that during normal prostate development Notch/PTEN pathway functions to maintain patterned progenitors in the epithelial and smooth muscle compartments. In addition, we found that both positive and negative modulation of Notch signaling results in abnormal organization of the prostate tissue, and can contribute to prostate disease in the adult organ.
Collapse
|
94
|
Barve A, Khor TO, Reuhl K, Reddy B, Newmark H, Kong AN. Mixed tocotrienols inhibit prostate carcinogenesis in TRAMP mice. Nutr Cancer 2010; 62:789-94. [PMID: 20661828 DOI: 10.1080/01635581003605896] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The biological activities of tocotrienols are receiving increasing attention. Herein, we report the efficacy of a mixed-tocotrienol diet against prostate tumorigenesis in the transgenic adenocarcinoma mouse prostate (TRAMP) mouse model. Male TRAMP mice, 8 wk old, were fed 0.1%, 0.3%, or 1% mixed tocotrienols in AIN-76A diet up to 24 wk old. Likewise, a positive control group consisting of male TRAMP mice and a negative control group consisting of wild-type nontransgenic mice were fed regular AIN-76A diet up to 24 wk old. Our results show that mixed-tocotrienol-fed groups had a lower incidence of tumor formation along with a significant reduction in the average wet weight of genitourinary apparatus. Furthermore, mixed tocotrienols significantly reduced the levels of high-grade neoplastic lesions as compared to the positive controls. This decrease in levels of high-grade neoplastic lesions was found to be associated with increased expression of proapoptotic proteins BAD (Bcl(2) antagonist of cell death) and cleaved caspase-3 and cell cycle regulatory proteins cyclin dependent kinase inhibitors p21 and p27. In contrast, the expression of cyclins A and E were found to be decreased in mixed-tocotrienol groups. Taken together, our results show that by modulating cell cycle regulatory proteins and increasing expression of proapoptotic proteins, mixed tocotrienols suppress prostate tumorigenesis in the TRAMP mice.
Collapse
Affiliation(s)
- Avantika Barve
- Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
95
|
Local prolactin is a target to prevent expansion of basal/stem cells in prostate tumors. Proc Natl Acad Sci U S A 2010; 107:15199-204. [PMID: 20699217 DOI: 10.1073/pnas.0911651107] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Androgen-independent recurrence is the major limit of androgen ablation therapy for prostate cancer. Identification of alternative pathways promoting prostate tumor growth is thus needed. Stat5 has been recently shown to promote human prostate cancer cell survival/proliferation and to be associated with early prostate cancer recurrence. Stat5 is the main signaling pathway triggered by prolactin (PRL), a growth factor whose local production is also increased in high-grade prostate cancers. The first aim of this study was to use prostate-specific PRL transgenic mice to address the mechanisms by which local PRL induces prostate tumorogenesis. We report that (i) Stat5 is the major signaling cascade triggered by local PRL in the mouse dorsal prostate, (ii) this model recapitulates prostate tumorogenesis from precancer lesions to invasive carcinoma, and (iii) tumorogenesis involves dramatic accumulation and abnormal spreading of p63-positive basal cells, and of stem cell antigen-1-positive cells identified as a stem/progenitor-like subpopulation. Because basal epithelial stem cells are proposed to serve as tumor-initiating cells, we challenged the relevance of local PRL as a previously unexplored therapeutic target. Using a double-transgenic approach, we show that Delta1-9-G129R-hPRL, a competitive PRL-receptor antagonist, prevented early stages of prostate tumorogenesis by reducing or inhibiting Stat5 activation, cell proliferation, abnormal basal-cell pattern, and frequency or grade of intraepithelial neoplasia. This study identifies PRL receptor/Stat5 as a unique pathway, initiating prostate tumorogenesis by altering basal-/stem-like cell subpopulations, and strongly supports the importance of further developing strategies to target locally overexpressed PRL in human prostate cancer.
Collapse
|
96
|
eIF4E phosphorylation promotes tumorigenesis and is associated with prostate cancer progression. Proc Natl Acad Sci U S A 2010; 107:14134-9. [PMID: 20679199 DOI: 10.1073/pnas.1005320107] [Citation(s) in RCA: 403] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Translational regulation plays a critical role in the control of cell growth and proliferation. A key player in translational control is eIF4E, the mRNA 5' cap-binding protein. Aberrant expression of eIF4E promotes tumorigenesis and has been implicated in cancer development and progression. The activity of eIF4E is dysregulated in cancer. Regulation of eIF4E is partly achieved through phosphorylation. However, the physiological significance of eIF4E phosphorylation in mammals is not clear. Here, we show that knock-in mice expressing a nonphosphorylatable form of eIF4E are resistant to tumorigenesis in a prostate cancer model. By using a genome-wide analysis of translated mRNAs, we show that the phosphorylation of eIF4E is required for translational up-regulation of several proteins implicated in tumorigenesis. Accordingly, increased phospho-eIF4E levels correlate with disease progression in patients with prostate cancer. Our findings establish eIF4E phosphorylation as a critical event in tumorigenesis. These findings raise the possibility that chemical compounds that prevent the phosphorylation of eIF4E could act as anticancer drugs.
Collapse
|
97
|
Francis JC, McCarthy A, Thomsen MK, Ashworth A, Swain A. Brca2 and Trp53 deficiency cooperate in the progression of mouse prostate tumourigenesis. PLoS Genet 2010; 6:e1000995. [PMID: 20585617 PMCID: PMC2891704 DOI: 10.1371/journal.pgen.1000995] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Accepted: 05/19/2010] [Indexed: 01/07/2023] Open
Abstract
Epidemiological studies have shown that one of the strongest risk factors for prostate cancer is a family history of the disease, suggesting that inherited factors play a major role in prostate cancer susceptibility. Germline mutations in BRCA2 predispose to breast and ovarian cancer with its predominant tumour suppressor function thought to be the repair of DNA double-strand breaks. BRCA2 has also been implicated in prostate cancer etiology, but it is unclear the impact that mutations in this gene have on prostate tumourigenesis. Here we have undertaken a genetic analysis in the mouse to determine the role of Brca2 in the adult prostate. We show that deletion of Brca2 specifically in prostate epithelia results in focal hyperplasia and low-grade prostate intraepithelial neoplasia (PIN) in animals over 12 months of age. Simultaneous deletion of Brca2 and the tumour suppressor Trp53 in prostate epithelia gave rise to focal hyperplasia and atypical cells at 6 months, leading to high-grade PIN in animals from 12 months. Epithelial cells in these lesions show an increase in DNA damage and have higher levels of proliferation, but also elevated apoptosis. Castration of Brca2;Trp53 mutant animals led to regression of PIN lesions, but atypical cells persisted that continued to proliferate and express nuclear androgen receptor. This study provides evidence that Brca2 can act as a tumour suppressor in the prostate, and the model we describe should prove useful in the development of new therapeutic approaches. In Western countries, prostate cancer is the most common male cancer and the second biggest cause of cancer-related deaths in men. Men with a familial history of either breast or ovarian cancer have an elevated predisposition to prostate cancer, suggesting there is a genetic element to this disease. Indeed, the inheritance of a mutated form of the breast cancer susceptibility gene BRCA2 has been linked to the development of prostate cancer, although the precise role that BRCA2 dysfunction plays in the development of prostate cancer is unclear. To address this, we have generated an animal model in which the mouse Brca2 gene is specifically deleted in the adult prostate. These mice develop precancerous prostate lesions, which progress in severity and incidence with the loss-of-function of an additional tumour suppressor, Trp53. Importantly, blocking male steroidal hormone production by castration leads to partial regression of the prostate lesions, however cells continue to proliferate after androgen withdrawal. This suggests human BRCA2 mutant prostate tumours, like the majority of prostate cancers, will respond to hormone therapy, but will relapse, as frequently occurs in this disease. In summary, our model suggests that BRCA2 acts as a tumour suppressor in the prostate and provides a pre-invasive model to test novel therapeutics.
Collapse
Affiliation(s)
- Jeffrey C. Francis
- Section of Gene Function and Regulation, Institute of Cancer Research, London, United Kingdom
| | - Afshan McCarthy
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Martin K. Thomsen
- Section of Gene Function and Regulation, Institute of Cancer Research, London, United Kingdom
| | - Alan Ashworth
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Amanda Swain
- Section of Gene Function and Regulation, Institute of Cancer Research, London, United Kingdom
- * E-mail:
| |
Collapse
|
98
|
Iwata T, Schultz D, Hicks J, Hubbard GK, Mutton LN, Lotan TL, Bethel C, Lotz MT, Yegnasubramanian S, Nelson WG, Dang CV, Xu M, Anele U, Koh CM, Bieberich CJ, De Marzo AM. MYC overexpression induces prostatic intraepithelial neoplasia and loss of Nkx3.1 in mouse luminal epithelial cells. PLoS One 2010; 5:e9427. [PMID: 20195545 PMCID: PMC2828486 DOI: 10.1371/journal.pone.0009427] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 01/26/2010] [Indexed: 12/12/2022] Open
Abstract
Lo-MYC and Hi-MYC mice develop prostatic intraepithelial neoplasia (PIN) and prostatic adenocarcinoma as a result of MYC overexpression in the mouse prostate. However, prior studies have not determined precisely when, and in which cell types, MYC is induced. Using immunohistochemistry (IHC) to localize MYC expression in Lo-MYC transgenic mice, we show that morphological and molecular alterations characteristic of high grade PIN arise in luminal epithelial cells as soon as MYC overexpression is detected. These changes include increased nuclear and nucleolar size and large scale chromatin remodeling. Mouse PIN cells retained a columnar architecture and abundant cytoplasm and appeared as either a single layer of neoplastic cells or as pseudo-stratified/multilayered structures with open glandular lumina-features highly analogous to human high grade PIN. Also using IHC, we show that the onset of MYC overexpression and PIN development coincided precisely with decreased expression of the homeodomain transcription factor and tumor suppressor, Nkx3.1. Virtually all normal appearing prostate luminal cells expressed high levels of Nkx3.1, but all cells expressing MYC in PIN lesions showed marked reductions in Nkx3.1, implicating MYC as a key factor that represses Nkx3.1 in PIN lesions. To determine the effects of less pronounced overexpression of MYC we generated a new line of mice expressing MYC in the prostate under the transcriptional control of the mouse Nkx3.1 control region. These "Super-Lo-MYC" mice also developed PIN, albeit a less aggressive form. We also identified a histologically defined intermediate step in the progression of mouse PIN into invasive adenocarcinoma. These lesions are characterized by a loss of cell polarity, multi-layering, and cribriform formation, and by a "paradoxical" increase in Nkx3.1 protein. Similar histopathological changes occurred in Hi-MYC mice, albeit with accelerated kinetics. Our results using IHC provide novel insights that support the contention that MYC overexpression is sufficient to transform prostate luminal epithelial cells into PIN cells in vivo. We also identified a novel histopathologically identifiable intermediate step prior to invasion that should facilitate studies of molecular pathway alterations occurring during early progression of prostatic adenocarcinomas.
Collapse
Affiliation(s)
- Tsuyoshi Iwata
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Denise Schultz
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jessica Hicks
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Gretchen K. Hubbard
- Department of Biological Sciences, The University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Laura N. Mutton
- Department of Biological Sciences, The University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Tamara L. Lotan
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Carlise Bethel
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew T. Lotz
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Srinivasan Yegnasubramanian
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William G. Nelson
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chi V. Dang
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Hematology and Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - MengMeng Xu
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Uzoma Anele
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Cheryl M. Koh
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Charles J. Bieberich
- Department of Biological Sciences, The University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Angelo M. De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
99
|
Thomsen MK, Ambroisine L, Wynn S, Cheah KSE, Foster CS, Fisher G, Berney DM, Møller H, Reuter VE, Scardino P, Cuzick J, Ragavan N, Singh PB, Martin FL, Butler CM, Cooper CS, Swain A. SOX9 elevation in the prostate promotes proliferation and cooperates with PTEN loss to drive tumor formation. Cancer Res 2010; 70:979-87. [PMID: 20103652 DOI: 10.1158/0008-5472.can-09-2370] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dysregulation of tissue development pathways can contribute to cancer initiation and progression. In murine embryonic prostate epithelia, the transcription factor SOX9 is required for proper prostate development. In this study, we examined a role for SOX9 in prostate cancer in mouse and human. In Pten and Nkx3.1 mutant mice, cells with increased levels of SOX9 appeared within prostate epithelia at early stages of neoplasia, and higher expression correlated with progression at all stages of disease. In transgenic mice, SOX9 overexpression in prostate epithelia increased cell proliferation without inducing hyperplasia. In transgenic mice that were also heterozygous for mutant Pten, SOX9 overexpression quickened the induction of high-grade prostate intraepithelial neoplasia. In contrast, Sox9 attenuation led to a decrease proliferating prostate epithelia cells in normal and homozygous Pten mutant mice with prostate neoplasia. Analysis of a cohort of 880 human prostate cancer samples showed that SOX9 expression was associated with increasing Gleason grades and higher Ki67 staining. Our findings identify SOX9 as part of a developmental pathway that is reactivated in prostate neoplasia where it promotes tumor cell proliferation.
Collapse
Affiliation(s)
- Martin K Thomsen
- Section of Gene Function and Regulation, Institute of Cancer Research, London SW3 6JB, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Recognition of focal morphological intraepithelial lesions associated with the eventual development of invasive cancer has long been the sine qua non of precancer. Empirically, precancers are associated with a morphological continuum from atypia to dysplasia and invasive neoplasia. Such lesions are used as early indicators of cancers and have dramatically reduced mortality from cancers of the colon, uterine cervix, and breast. Progression has been modeled as a linear, stepwise process. Some molecular evidence supports a linear model. However, clinical studies now suggest that preexisting cofactors such as human papilloma virus (HPV) in cervical cancer determines the cell fate. Other clinical studies such as bladder, prostate, and breast suggest that many intraepithelial lesions do not progress to malignancy. The more recent experimental analyses reveal that the key molecular and genetic events even predate the emergence of visible lesions. Thus, a new nonlinear, parallel model is proposed. The parallel model suggests an origin in a putative progenitor cell that expands and invades. The clinical outcome is thus predetermined. If correct, this model suggests that "progression" to malignancy is epigenetic. Further, future assessment of biological potential will involve identification and genetic analysis of the progenitor cell populations.
Collapse
|