101
|
Abstract
The clinical course of prostate cancer is grouped into two broad phases. The first phase, which is the growth of the androgen-dependent cancer (AD-Ca) responds well to androgen depletion treatment while the second phase, that could be termed as androgen depletion-independent cancer (ADI-Ca) does not. We used two separate prostate tumors, one AD-Ca and one ADI-Ca from the conditional Pten deletion mouse model to generate from each a pair of cell lines. The AD-Ca cell lines (E2 and E4) and the ADI-Ca cell lines (cE1 and cE2) display bi-allelic deletion at the Pten gene locus, an event which is specific for the prostate epithelium for this mouse model, and a fairly similar level of expression of the androgen receptor (AR). The ADI-Ca cell lines (cE series) grow well in the absence of androgen, display increased AR transcription under androgen-deprived environment, and retain the sensitivity to increased proliferation when androgen is supplemented. The AD-Ca cell lines (E series) grow slowly in the absence of androgen, and, unlike cE cells, do not show increased AR expression when maintained in the absence of androgen. The detection of epithelial cell markers, such as CK8, CK14, CK18 and E-cadherin in the cE series is conforming with the polygonal epithelial morphology of these cells in culture. The E cells also present mostly polygonal-shaped morphology with a small percent of cells with fibroblastoid morphology, and produce little or very low levels of cytokeratins, but increased levels of vimentin, Twist and Slug, the markers known to be associated with epithelial-mesenchymal transition. Each of the cell lines, when inoculated subcutaneously into male or female NOD.SCID mice induced tumors within eight weeks with 100% incidence. Histopathological examinations of the tumor sections, however, led to noticeable biological differences. The cE series engenders adenocarcinomas, particularly in male hosts, and the E series induces sarcomatoid carcinomas (positively stained for CK8 and AR as well as vimentin expression) in either male or female hosts. These new cell lines are promising models for the elucidation of the androgen metabolism and their role in prostate cancer.
Collapse
|
102
|
Lai KP, Yamashita S, Vitkus S, Shyr CR, Yeh S, Chang C. Suppressed prostate epithelial development with impaired branching morphogenesis in mice lacking stromal fibromuscular androgen receptor. Mol Endocrinol 2011; 26:52-66. [PMID: 22135068 DOI: 10.1210/me.2011-1189] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Using the cre-loxP system, we generated a new mouse model [double stromal androgen receptor knockout (dARKO)] with selectively deleted androgen receptor (AR) in both stromal fibroblasts and smooth muscle cells, and found the size of the anterior prostate (AP) lobes was significantly reduced as compared with those from wild-type littermate controls. The reduction in prostate size of the dARKO mouse was accompanied by impaired branching morphogenesis and partial loss of the infolding glandular structure. Further dissection found decreased proliferation and increased apoptosis of the prostate epithelium in the dARKO mouse AP. These phenotype changes were further confirmed with newly established immortalized prostate stromal cells (PrSC) from wild-type and dARKO mice. Mechanistically, IGF-1, placental growth factor, and secreted phosphoprotein-1 controlled by stromal AR were differentially expressed in PrSC-wt and PrSC-ARKO. Moreover, the conditioned media (CM) from PrSC-wt promoted prostate epithelium growth significantly as compared with CM from PrSC-dARKO. Finally, adding IGF-1/placental growth factor recombinant proteins into PrSC-dARKO CM was able to partially rescue epithelium growth. Together, our data concluded that stromal fibromuscular AR could modulate epithelium growth and maintain cellular homeostasis through identified growth factors.
Collapse
Affiliation(s)
- Kuo-Pao Lai
- George H Whipple Laboratory for Cancer Research, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | |
Collapse
|
103
|
Abstract
Two recent Cancer Cell articles report the discovery of reciprocal feedback regulation between androgenic and PTEN loss/PI3K-AKT signaling in prostate cancer. Both studies link endocrine regulation with a common oncogenic pathway, which led to the development of a combination therapeutic approach with immediate application in prostate cancer.
Collapse
Affiliation(s)
- Timothy C Thompson
- Department of Genitourinary Medical Oncology-Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA.
| |
Collapse
|
104
|
Mulholland DJ, Tran LM, Li Y, Cai H, Morim A, Wang S, Plaisier S, Garraway IP, Huang J, Graeber TG, Wu H. Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth. Cancer Cell 2011; 19:792-804. [PMID: 21620777 PMCID: PMC3157296 DOI: 10.1016/j.ccr.2011.05.006] [Citation(s) in RCA: 404] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 04/04/2011] [Accepted: 05/05/2011] [Indexed: 12/21/2022]
Abstract
Alteration of the PTEN/PI3K pathway is associated with late-stage and castrate-resistant prostate cancer (CRPC). However, how PTEN loss is involved in CRPC development is not clear. Here, we show that castration-resistant growth is an intrinsic property of Pten null prostate cancer (CaP) cells, independent of cancer development stage. PTEN loss suppresses androgen-responsive gene expressions by modulating androgen receptor (AR) transcription factor activity. Conditional deletion of Ar in the epithelium promotes the proliferation of Pten null cancer cells, at least in part, by downregulating the androgen-responsive gene Fkbp5 and preventing PHLPP-mediated AKT inhibition. Our findings identify PI3K and AR pathway crosstalk as a mechanism of CRPC development, with potentially important implications for CaP etiology and therapy.
Collapse
Affiliation(s)
- David J Mulholland
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Linh M. Tran
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Yunfeng Li
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Houjian Cai
- Department of Microbiology, Immunology and Molecular Medicine, University of California, Los Angeles
| | - Ashkan Morim
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Shunyou Wang
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Seema Plaisier
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | | | - Jiaoti Huang
- Department of Pathology, University of California, Los Angeles
| | - Thomas G. Graeber
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
| | - Hong Wu
- Department of Molecular and Medical Pharmacology and Institute for Molecular Medicine, University of California, Los Angeles
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles
- Corresponding author: Hong Wu, M.D, Ph.D. 650 Charles E Young Drive S. Los Angeles, CA 90095, Phone: 310.825.5160, FAX: 310.267.0242,
| |
Collapse
|
105
|
Wang X, Raulji P, Mohapatra SS, Patel R, Hellermann G, Kong X, Vera PL, Meyer-Siegler KL, Coppola D, Mohapatra S. Natriuretic peptide receptor a as a novel target for prostate cancer. Mol Cancer 2011; 10:56. [PMID: 21586128 PMCID: PMC3121714 DOI: 10.1186/1476-4598-10-56] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 05/17/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The receptor for the cardiac hormone atrial natriuretic peptide (ANP), natriuretic peptide receptor A (NPRA), is expressed in cancer cells, and natriuretic peptides have been implicated in cancers. However, the direct role of NPRA signaling in prostate cancer remains unclear. RESULTS NPRA expression was examined by western blotting, RT-PCR and immunohistochemistry. NPRA was downregulated by transfection of siRNA, shRNA and NPRA inhibitor (iNPRA). Antitumor efficacy of iNPRA was tested in mice using a TRAMP-C1 xenograft. Here, we demonstrated that NPRA is abundantly expressed on tumorigenic mouse and human prostate cells, but not in nontumorigenic prostate epithelial cells. NPRA expression showed positive correlation with clinical staging in a human PCa tissue microarray. Down-regulation of NPRA by siNPRA or iNPRA induced apoptosis in PCa cells. The mechanism of iNPRA-induced anti-PCa effects was linked to NPRA-induced expression of macrophage migration inhibitory factor (MIF), a proinflammatory cytokine over-expressed in PCa and significantly reduced by siNPRA. Prostate tumor cells implanted in mice deficient in atrial natriuretic peptide receptor A (NPRA-KO) failed to grow, and treatment of TRAMP-C1 xenografts with iNPRA reduced tumor burden and MIF expression. Using the TRAMP spontaneous PCa model, we found that NPRA expression correlated with MIF expression during PCa progression. CONCLUSIONS Collectively, these results suggest that NPRA promotes PCa development in part by regulating MIF. Our findings also suggest that NPRA is a potential prognostic marker and a target for PCa therapy.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, Arora VK, Le C, Koutcher J, Scher H, Scardino PT, Rosen N, Sawyers CL. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 2011; 19:575-86. [PMID: 21575859 PMCID: PMC3142785 DOI: 10.1016/j.ccr.2011.04.008] [Citation(s) in RCA: 925] [Impact Index Per Article: 71.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 02/24/2011] [Accepted: 04/14/2011] [Indexed: 12/15/2022]
Abstract
Prostate cancer is characterized by its dependence on androgen receptor (AR) and frequent activation of PI3K signaling. We find that AR transcriptional output is decreased in human and murine tumors with PTEN deletion and that PI3K pathway inhibition activates AR signaling by relieving feedback inhibition of HER kinases. Similarly, AR inhibition activates AKT signaling by reducing levels of the AKT phosphatase PHLPP. Thus, these two oncogenic pathways cross-regulate each other by reciprocal feedback. Inhibition of one activates the other, thereby maintaining tumor cell survival. However, combined pharmacologic inhibition of PI3K and AR signaling caused near-complete prostate cancer regressions in a Pten-deficient murine prostate cancer model and in human prostate cancer xenografts, indicating that both pathways coordinately support survival.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Feedback, Physiological
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Humans
- Magnetic Resonance Imaging
- Male
- Mice
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Nuclear Proteins/metabolism
- PTEN Phosphohydrolase/deficiency
- PTEN Phosphohydrolase/genetics
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Phosphoprotein Phosphatases/metabolism
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- RNA Interference
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/metabolism
- Receptors, Androgen/drug effects
- Receptors, Androgen/metabolism
- Signal Transduction/drug effects
- Time Factors
- Transcription, Genetic
- Transfection
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Brett S Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Surgery and Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Caren Chapinski
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Surgery and Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - John Wongvipat
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Haley Hieronymus
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Vivek K Arora
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Carl Le
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jason Koutcher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Howard Scher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Peter T Scardino
- Department of Surgery and Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Neal Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Molecular Pharmacology and Chemistry, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
107
|
Role of autonomous androgen receptor signaling in prostate cancer initiation is dichotomous and depends on the oncogenic signal. Proc Natl Acad Sci U S A 2011; 108:7962-7. [PMID: 21518863 DOI: 10.1073/pnas.1105243108] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The steroid hormone signaling axis is thought to play a central role in initiation and progression of many hormonally regulated epithelial tumors. It is unclear whether all cancer-initiating signals depend on an intact hormone receptor signaling machinery. To ascertain whether cell autonomous androgen receptor (AR) is essential for initiation of prostate intraepithelial neoplasia (PIN), the response of AR-null prostate epithelia to paracrine and cell autonomous oncogenic signals was assessed in vivo by using the prostate regeneration model system. Epithelial-specific loss of AR blocked paracrine FGF10-induced PIN, whereas the add back of exogenous AR restored this response. In contrast, PIN initiated by cell-autonomous, chronic-activated AKT developed independent of epithelial AR signaling. Our findings demonstrate a selective role for AR in the initiation of PIN, dependent on the signaling pathways driving tumor formation. Insights into the role of hormone receptor signaling in the initiation of epithelial tumors may help define this axis as a target for chemoprevention of carcinomas.
Collapse
|
108
|
Aggarwal R, Ryan CJ. Castration-resistant prostate cancer: targeted therapies and individualized treatment. Oncologist 2011; 16:264-75. [PMID: 21339259 PMCID: PMC3228103 DOI: 10.1634/theoncologist.2010-0216] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 01/14/2011] [Indexed: 01/14/2023] Open
Abstract
Various molecular mechanisms have been implicated in the progression from hormone-sensitive to castration-resistant prostate cancer (CRPC). Novel targeted agents to treat CRPC have been developed that inhibit either androgen receptor (AR)-mediated signaling (AR antagonists and inhibitors of androgen synthesis) or non-AR-mediated signaling (inhibitors of Src, mammalian target of rapamycin, chaperone proteins, insulin-like growth factor-1 receptor, vascular endothelial growth factor, and endothelin-A receptor) pathways. However, variable efficacy has been observed in clinical trials, most likely because of the biologic heterogeneity of CRPC. To account for potential differences in disease biology, a more individualized approach to treatment, based on genomic and/or proteomic analyses of individual tumors, is being investigated. By identifying tumors with a characteristic molecular subtype and assigning treatment accordingly, it is hoped that a higher proportion of patients will benefit from targeted therapy. Additionally, lessons learned through the application of these technologies to prostate cancer may subsequently influence therapeutic development in other solid tumors.
Collapse
Affiliation(s)
- Rahul Aggarwal
- Department of Medicine, University of California, San Francisco, CA 94143-1270, USA.
| | | |
Collapse
|
109
|
Modeling Human Prostate Cancer in Genetically Engineered Mice. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:1-49. [DOI: 10.1016/b978-0-12-384878-9.00001-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
110
|
Shen MM, Abate-Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev 2010; 24:1967-2000. [PMID: 20844012 DOI: 10.1101/gad.1965810] [Citation(s) in RCA: 693] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite much recent progress, prostate cancer continues to represent a major cause of cancer-related mortality and morbidity in men. Since early studies on the role of the androgen receptor that led to the advent of androgen deprivation therapy in the 1940s, there has long been intensive interest in the basic mechanisms underlying prostate cancer initiation and progression, as well as the potential to target these processes for therapeutic intervention. Here, we present an overview of major themes in prostate cancer research, focusing on current knowledge of principal events in cancer initiation and progression. We discuss recent advances, including new insights into the mechanisms of castration resistance, identification of stem cells and tumor-initiating cells, and development of mouse models for preclinical evaluation of novel therapuetics. Overall, we highlight the tremendous research progress made in recent years, and underscore the challenges that lie ahead.
Collapse
Affiliation(s)
- Michael M Shen
- Department of Medicine, Columbia University Medical Center, New York, New York 10032, USA.
| | | |
Collapse
|
111
|
Bohrer LR, Chen S, Hallstrom TC, Huang H. Androgens suppress EZH2 expression via retinoblastoma (RB) and p130-dependent pathways: a potential mechanism of androgen-refractory progression of prostate cancer. Endocrinology 2010; 151:5136-45. [PMID: 20881251 PMCID: PMC2954725 DOI: 10.1210/en.2010-0436] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Androgens and the androgen receptor are important for both normal prostate development and progression of prostate cancer (PCa). However, the underlying mechanisms are not fully understood. The Polycomb protein enhancer of zeste homolog 2 (EZH2) functions as an epigenetic gene silencer and plays a role in oncogenesis by promoting cell proliferation and invasion. EZH2 has been implicated in human PCa progression, because its expression is often elevated in hormone-refractory PCa. Here, we demonstrated that expression of EZH2 is lower in androgen-sensitive LNCaP PCa cells compared with Rf and C4-2 cells, two androgen-refractory sublines that are derived from LNCaP cells. Androgen ablation by castration increased the level of EZH2 proteins in LNCaP xenografts in mice. In contrast, treatment of LNCaP cells in culture with the synthetic androgen methyltrieolone (R1881) at doses of 1 nm or higher suppressed EZH2 expression. Moreover, our data suggest that androgen repression of EZH2 requires a functional androgen receptor and this effect is mediated through the retinoblastoma protein and its related protein p130. We further showed that androgen treatment not only increases expression of EZH2 target genes DAB2IP and E-cadherin but also affects LNCaP cell migration. Our results reveal that androgens function as an epigenetic regulator in prostatic cells by repression of EZH2 expression through the retinoblastoma protein and p130-dependent pathways. Our findings also suggest that blockade of EZH2 derepression during androgen deprivation therapy may represent an effective tactic for the treatment of androgen-refractory PCa.
Collapse
Affiliation(s)
- Laura R Bohrer
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
112
|
Plaisier SB, Taschereau R, Wong JA, Graeber TG. Rank-rank hypergeometric overlap: identification of statistically significant overlap between gene-expression signatures. Nucleic Acids Res 2010; 38:e169. [PMID: 20660011 PMCID: PMC2943622 DOI: 10.1093/nar/gkq636] [Citation(s) in RCA: 313] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Comparing independent high-throughput gene-expression experiments can generate hypotheses about which gene-expression programs are shared between particular biological processes. Current techniques to compare expression profiles typically involve choosing a fixed differential expression threshold to summarize results, potentially reducing sensitivity to small but concordant changes. We present a threshold-free algorithm called Rank–rank Hypergeometric Overlap (RRHO). This algorithm steps through two gene lists ranked by the degree of differential expression observed in two profiling experiments, successively measuring the statistical significance of the number of overlapping genes. The output is a graphical map that shows the strength, pattern and bounds of correlation between two expression profiles. To demonstrate RRHO sensitivity and dynamic range, we identified shared expression networks in cancer microarray profiles driving tumor progression, stem cell properties and response to targeted kinase inhibition. We demonstrate how RRHO can be used to determine which model system or drug treatment best reflects a particular biological or disease response. The threshold-free and graphical aspects of RRHO complement other rank-based approaches such as Gene Set Enrichment Analysis (GSEA), for which RRHO is a 2D analog. Rank–rank overlap analysis is a sensitive, robust and web-accessible method for detecting and visualizing overlap trends between two complete, continuous gene-expression profiles. A web-based implementation of RRHO can be accessed at http://systems.crump.ucla.edu/rankrank/.
Collapse
Affiliation(s)
- Seema B Plaisier
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, Institute for Molecular Medicine, Jonsson Comprehensive Cancer Center and California NanoSystems Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
113
|
Suh Y, Afaq F, Khan N, Johnson JJ, Khusro FH, Mukhtar H. Fisetin induces autophagic cell death through suppression of mTOR signaling pathway in prostate cancer cells. Carcinogenesis 2010; 31:1424-33. [PMID: 20530556 DOI: 10.1093/carcin/bgq115] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) kinase is an important component of PTEN/PI3K/Akt signaling pathway, which is frequently deregulated in prostate cancer (CaP). Recent studies suggest that targeting PTEN/PI3K/Akt and mTOR signaling pathway could be an effective strategy for the treatment of hormone refractory CaP. Here, we show that the treatment of androgen-independent and PTEN-negative human CaP PC3 cells with fisetin, a dietary flavonoid, resulted in inhibition of mTOR kinase signaling pathway. Treatment of cells with fisetin inhibited mTOR activity and downregulated Raptor, Rictor, PRAS40 and GbetaL that resulted in loss of mTOR complexes (mTORC)1/2 formation. Fisetin also activated the mTOR repressor TSC2 through inhibition of Akt and activation of AMPK. Fisetin-mediated inhibition of mTOR resulted in hypophosphorylation of 4EBP1 and suppression of Cap-dependent translation. We also found that fisetin treatment leads to induction of autophagic-programmed cell death rather than cytoprotective autophagy as shown by small interfering RNA Beclin1-knockdown and autophagy inhibitor. Taken together, we provide evidence that fisetin functions as a dual inhibitor of mTORC1/2 signaling leading to inhibition of Cap-dependent translation and induction of autophagic cell death in PC3 cells. These results suggest that fisetin could be a useful chemotherapeutic agent in treatment of hormone refractory CaP.
Collapse
Affiliation(s)
- Yewseok Suh
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
114
|
Akins EJ, Moore ML, Tang S, Willingham MC, Tooze JA, Dubey P. In situ vaccination combined with androgen ablation and regulatory T-cell depletion reduces castration-resistant tumor burden in prostate-specific pten knockout mice. Cancer Res 2010; 70:3473-82. [PMID: 20406970 DOI: 10.1158/0008-5472.can-09-2490] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is no effective treatment for prostate cancer arising after androgen ablation. Previous studies have analyzed the short-term effects of androgen ablation on the immune system and suggest an abatement of immune suppression by hormone removal. Because castration-resistant disease can arise years after treatment, it is crucial to determine the duration of immune potentiation by castration. Because immunotherapeutic efficacy is determined by the balance of immune cell subsets and their location within the tumor, we assessed the acute and chronic effect of androgen ablation on the localization of T-cell subsets within castration-resistant murine prostate cancer. We observed a transient increase in CD4+ and CD8+ T-cell numbers at the residual tumor after androgen ablation. More than 2 months later, regulatory T cells (Treg) were increasingly found within prostate epithelium, whereas CTLs, which were evenly distributed before androgen ablation, became sequestered within stroma. Anti-CD25 antibody administration along with castration enhanced CTL access to cancerous glands but did not increase effector function. Intraprostatic injection of LIGHT-expressing tumor cells increased the proportion of CD8+ T cells with functional capacity within the cancerous gland. In addition, Treg depletion within the tumor was enhanced. Together, these manipulations significantly reduced castration-resistant tumor burden. Thus, our results indicate that immune modulations, which prevent Treg accumulation and augment effector cell infiltration of prostatic epithelium, may be effective in reducing tumor burden or preventing tumor recurrence after androgen ablation therapy.
Collapse
Affiliation(s)
- Elizabeth J Akins
- Department of Pathology-Tumor Biology, Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157-1092, USA
| | | | | | | | | | | |
Collapse
|
115
|
Sherman DJ, Kenanova VE, Lepin EJ, McCabe KE, Kamei KI, Ohashi M, Wang S, Tseng HR, Wu AM, Behrenbruch CP. A differential cell capture assay for evaluating antibody interactions with cell surface targets. Anal Biochem 2010; 401:173-81. [PMID: 20178770 DOI: 10.1016/j.ab.2010.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 01/24/2010] [Accepted: 02/13/2010] [Indexed: 11/26/2022]
Abstract
Many biological and biomedical laboratory assays require the use of antibodies and antibody fragments that strongly bind to their cell surface targets. Conventional binding assays, such as the enzyme-linked immunosorbent assay (ELISA) and flow cytometry, have many challenges, including capital equipment requirements, labor intensiveness, and large reagent and sample consumption. Although these techniques are successful in mainstream biology, there is an unmet need for a tool to quickly ascertain the relative binding capabilities of antibodies/antibody fragments to cell surface targets on the benchtop at low cost. We describe a novel cell capture assay that enables several candidate antibodies to be evaluated quickly as to their relative binding efficacies to their cell surface targets. We used chimeric rituximab and murine anti-CD20 monoclonal antibodies as cell capture agents on a functionalized microscope slide surface to assess their relative binding affinities based on how well they capture CD20-expressing mammalian cells. We found that these antibodies' concentration-dependent cell capture profiles correlate with their relative binding affinities. A key observation of this assay involved understanding how differences in capture surfaces affect the assay results. This approach can find utility when an antibody or antibody fragment against a known cell line needs to be selected for targeting studies.
Collapse
Affiliation(s)
- David J Sherman
- Department of Molecular and Medical Pharmacology, Crump Institute of Molecular Imaging, California NanoSystems Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Sircar K, Yoshimoto M, Monzon FA, Koumakpayi IH, Katz RL, Khanna A, Alvarez K, Chen G, Darnel AD, Aprikian AG, Saad F, Bismar TA, Squire JA. PTEN genomic deletion is associated with p-Akt and AR signalling in poorer outcome, hormone refractory prostate cancer. J Pathol 2009; 218:505-13. [PMID: 19402094 DOI: 10.1002/path.2559] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PTEN haploinsufficiency is common in hormone-sensitive prostate cancer, though the incidence of genomic deletion and its downstream effects have not been elucidated in clinical samples of hormone refractory prostate cancer (HRPC). Progression to androgen independence is pivotal in prostate cancer and mediated largely by the androgen receptor (AR). Since this process is distinct from metastatic progression, we examined alterations of the PTEN gene in locally advanced recurrent, non-metastatic human HRPC tissues. Retrospective analyses of PTEN deletion status were correlated with activated downstream phospho-Akt (p-Akt) pathway proteins and with the androgen receptor. The prevalence of PTEN genomic deletions in transurethral resection samples of 59 HRPC patients with known clinical outcome was assessed by four-colour FISH analyses. FISH was performed using six BAC clones spanning both flanking PTEN genomic regions and the PTEN gene locus, and a chromosome 10 centromeric probe. PTEN copy number was also evaluated in a subset of cases using single nucleotide polymorphism (SNP) arrays. In addition, the samples were immunostained with antibodies against p-Akt, p-mTOR, p-70S6, and AR. The PTEN gene was deleted in 77% of cases, with 25% showing homozygous deletions, 18% homozygous and hemizygous deletions, and 34% hemizygous deletions only. In a subset of the study group, SNP array analysis confirmed the FISH findings. PTEN genomic deletion was significantly correlated to the expression of downstream p-Akt (p < 0.0001), AR (p = 0.025), and to cancer-specific mortality (p = 0.039). PTEN deletion is common in HRPC, with bi-allelic loss correlating to disease-specific mortality and associated with Akt and AR deregulation.
Collapse
Affiliation(s)
- Kanishka Sircar
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Han B, Mehra R, Lonigro RJ, Wang L, Suleman K, Menon A, Palanisamy N, Tomlins SA, Chinnaiyan AM, Shah RB. Fluorescence in situ hybridization study shows association of PTEN deletion with ERG rearrangement during prostate cancer progression. Mod Pathol 2009; 22:1083-93. [PMID: 19407851 PMCID: PMC2760294 DOI: 10.1038/modpathol.2009.69] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The link between ERG rearrangement and PTEN (phosphatase and tensin homolog deleted on chromosome 10) deletion is unclear in prostate cancer progression. Using fluorescence in situ hybridization, we systematically validated the frequency and distribution of ERG and PTEN aberrations in a cohort of 73 benign prostate tissues, 59 high-grade prostatic intraepithelial neoplasia (HGPIN) foci, 281 localized prostate cancer and 47 androgen-independent metastatic prostate cancer patients. Overall, ERG rearrangement was present in 15% (5/33) of HGPIN, 45% (121/267) of localized cancers and 35% (15/43) of metastases. By contrast, PTEN deletion was identified in 9% (3/33) of HGPIN, 17% (42/251) of localized cancers and 54% (22/41) of metastases, of which 0%, 40% (17/42) and 45% (10/22) were homozygous, respectively. Concomitance of ERG rearrangement and PTEN deletion was observed in a subset of HGPIN. Significantly, association between PTEN deletion and ERG rearrangement was present both in localized cancers (P=0.0008) and metastases (P=0.02). Further, immunohistochemistry revealed significant correlation of decreased PTEN protein expression with PTEN genomic deletion both in localized and metastatic cancer. Of note, ERG aberration, but not PTEN deletion, was consistently identical both in localized cancer and adjacent HGPIN. Similarly, whereas all metastases (41/41, 100%) shared the same ERG status across multiple sites from the same patient, 5% (2/41) of cases showed discordance for PTEN deletion status across multiple sites. Collectively, our data support PTEN deletion as a late genetic event in human prostate cancer, presumably a 'second hit' after ERG rearrangement. PTEN deletion and ERG rearrangement may cooperate, but contribute at different stages, in prostate cancer progression.
Collapse
Affiliation(s)
- Bo Han
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Robert J. Lonigro
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Lei Wang
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Khalid Suleman
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Anjana Menon
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Nallasivam Palanisamy
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Scott A. Tomlins
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Rajal B. Shah
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan 48109,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
118
|
Narayanan NK, Nargi D, Randolph C, Narayanan BA. Liposome encapsulation of curcumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout mice. Int J Cancer 2009; 125:1-8. [PMID: 19326431 DOI: 10.1002/ijc.24336] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increasing interest in the use of phytochemicals to reduce prostate cancer led us to investigate 2 potential agents, curcumin and resveratrol as preventive agents. However, there is concern about the bioavailability of these agents pertinent to the poor absorption and thereby limiting its clinical use. With the view to improve their bioavailability, we used the liposome encapsulated curcumin, and resveratrol individually and in combination in male B6C3F1/J mice. Further, we examined the chemopreventive effect of liposome encapsulated curcumin and resveratrol in combination in prostate-specific PTEN knockout mice. In vitro assays using PTEN-CaP8 cancer cells were performed to investigate the combined effects curcumin with resveratrol on (i) cell growth, apoptosis and cell cycle (ii) impact on activated p-Akt, cyclin D1, m-TOR and androgen receptor (AR) proteins involved in tumor progression. HPLC analysis of serum and prostate tissues showed a significant increase in curcumin level when liposome encapsulated curcumin coadministered with liposomal resveratrol (p < 0.001). Combination of liposomal forms of curcumin and resveratrol significantly decreased prostatic adenocarcinoma in vivo (p < 0.001). In vitro studies revealed that curcumin plus resveratrol effectively inhibit cell growth and induced apoptosis. Molecular targets activated due to the loss of phosphatase and tensin homolog (PTEN) including p-Akt, cyclin D1, mammalian target of rapamycin and AR were downregulated by these agents in combination. Findings from this study for the first time provide evidence on phytochemicals in combination to enhance chemopreventive efficacy in prostate cancer. These findings clearly suggest that phytochemicals in combination may reduce prostate cancer incidence due to the loss of the tumor suppressor gene PTEN.
Collapse
Affiliation(s)
- Narayanan K Narayanan
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| | | | | | | |
Collapse
|
119
|
Simultaneous inactivation of Par-4 and PTEN in vivo leads to synergistic NF-kappaB activation and invasive prostate carcinoma. Proc Natl Acad Sci U S A 2009; 106:12962-7. [PMID: 19470463 DOI: 10.1073/pnas.0813055106] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer is one of the most common neoplasias in men. The tumor suppressor Par-4 is an important negative regulator of the canonical NF-kappaB pathway and is highly expressed in prostate. Here we show that Par-4 expression is lost in a high percentage of human prostate carcinomas, and this occurs in association with phosphatase and tensin homolog deleted from chromosome 10 (PTEN) loss. Par-4 null mice, similar to PTEN-heterozygous mice, only develop benign prostate lesions, but, importantly, concomitant Par-4 ablation and PTEN-heterozygosity lead to invasive prostate carcinoma in mice. This strong tumorigenic cooperation is anticipated in the preneoplastic prostate epithelium by an additive increase in Akt activation and a synergistic stimulation of NF-kappaB. These results establish the cooperation between Par-4 and PTEN as relevant for the development of prostate cancer and implicate the NF-kappaB pathway as a critical event in prostate tumorigenesis.
Collapse
|
120
|
Lafferty-Whyte K, Cairney CJ, Jamieson NB, Oien KA, Keith WN. Pathway analysis of senescence-associated miRNA targets reveals common processes to different senescence induction mechanisms. Biochim Biophys Acta Mol Basis Dis 2009; 1792:341-52. [DOI: 10.1016/j.bbadis.2009.02.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 02/02/2009] [Accepted: 02/03/2009] [Indexed: 01/07/2023]
|
121
|
Jones JO, An WF, Diamond MI. AR inhibitors identified by high-throughput microscopy detection of conformational change and subcellular localization. ACS Chem Biol 2009; 4:199-208. [PMID: 19236099 PMCID: PMC2776083 DOI: 10.1021/cb900024z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Signaling via the androgen receptor (AR) plays an important role in human health and disease. All currently available anti-androgens prevent ligand access to the receptor, either by limiting androgen synthesis or by competitive antagonism at the ligand binding domain. It is unknown to what extent various steps of receptor activation may be separable and distinctly targeted by inhibitors. We have previously described the use of fluorescent protein fusions to AR to monitor its subcellular distribution and ligand-induced conformational change by fluorescence resonance energy transfer (FRET). We have now used a microscopy-based screen to identify inhibitors that prevent AR conformational change or nuclear accumulation after ligand activation. Hits were secondarily selected on the basis of their ability to inhibit AR transcription at a PSA-luciferase promoter and were tested for effects on (3)H-DHT binding to AR in cells. We find a strong correlation between compounds that block DHT binding and those that inhibit nuclear accumulation. These compounds are structurally distinct from known antagonists. Additional compounds blocked AR conformational change but did not affect DHT binding or nuclear localization of AR. One compound increased ligand-induced FRET yet functioned as a potent inhibitor. These results suggest that multiple inhibitory conformations of AR are possible and can be induced by diverse mechanisms. The lead compounds described here may be candidates for the development of novel antiandrogens and may help identify new therapeutic targets.
Collapse
Affiliation(s)
- Jeremy O Jones
- Department of Neurology, UCSF, San Francisco, California, USA
| | | | | |
Collapse
|
122
|
Bratland A, Boender PJ, Høifødt HK, Østensen IHG, Ruijtenbeek R, Wang MY, Berg JP, Lilleby W, Fodstad Ø, Ree AH. Osteoblast-induced EGFR/ERBB2 signaling in androgen-sensitive prostate carcinoma cells characterized by multiplex kinase activity profiling. Clin Exp Metastasis 2009; 26:485-96. [PMID: 19294521 DOI: 10.1007/s10585-009-9248-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Accepted: 02/27/2009] [Indexed: 10/21/2022]
Abstract
Bone metastases in prostate cancer are predominantly osteoblastic. To study regulatory mechanisms underlying the establishment of prostate cancer within an osteoblastic microenvironment, human androgen-sensitive prostate carcinoma cells (LNCaP) were treated with culture medium conditioned by human osteoblast-derived sarcoma cells (OHS), and activated signalling pathways in the carcinoma cells were analyzed using microarrays with tyrosine kinase substrates. Network interaction analysis of substrates with significantly increased phosphorylation levels revealed that signalling pathways mediated by EGFR and ERBB2 were activated in LNCaP cells under OHS influence but also by androgen treatment. Activation of EGFR/ERBB2 signalling was also found in LNCaP cells in cocultures with OHS cells or osteoblastic cells that had been differentiated from human mesenchymal stem cells. Our experimental data suggests osteoblast-directed induction of signalling activity via EGFR and ERBB2 in prostate carcinoma cells and may provide a rationale for the use of EGFR or ERBB2 inhibition in systemic prevention or treatment of metastatic prostate cancer in the androgen-sensitive stage of the disease.
Collapse
Affiliation(s)
- Ase Bratland
- Department of Tumor Biology, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Fibroblast growth factor receptors (FGFRs) comprise a subfamily of receptor tyrosine kinases (RTKs) that are master regulators of a broad spectrum of cellular and developmental processes, including apoptosis, proliferation, migration and angiogenesis. Due to their broad impact, FGFRs and other RTKs are highly regulated and normally only basally active. Deregulation of FGFR signaling by activating mutations or ligand/receptor overexpression could allow these receptors to become constitutively active, leading to cancer development, including both hematopoietic and solid tumors, such as breast, bladder and prostate carcinomas. In this review, we focus on potential modes of FGFR-mediated tumorigenesis, in particular, the role of FGFR1 during prostate cancer progression.
Collapse
Affiliation(s)
- Victor D Acevedo
- Program in Cell and Molecular Biology, Department of Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
124
|
Guertin DA, Stevens DM, Saitoh M, Kinkel S, Crosby K, Sheen JH, Mullholland DJ, Magnuson MA, Wu H, Sabatini DM. mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell 2009; 15:148-59. [PMID: 19185849 PMCID: PMC2701381 DOI: 10.1016/j.ccr.2008.12.017] [Citation(s) in RCA: 311] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 10/22/2008] [Accepted: 12/18/2008] [Indexed: 12/21/2022]
Abstract
mTOR complex 2 (mTORC2) contains the mammalian target of rapamycin (mTOR) kinase and the Rictor regulatory protein and phosphorylates Akt. Whether this function of mTORC2 is critical for cancer progression is unknown. Here, we show that transformed human prostate epithelial cells lacking PTEN require mTORC2 to form tumors when injected into nude mice. Furthermore, we find that Rictor is a haploinsufficient gene and that deleting one copy protects Pten heterozygous mice from prostate cancer. Finally, we show that the development of prostate cancer caused by Pten deletion specifically in prostate epithelium requires mTORC2, but that for normal prostate epithelial cells, mTORC2 activity is nonessential. The selective requirement for mTORC2 in tumor development suggests that mTORC2 inhibitors may be of substantial clinical utility.
Collapse
Affiliation(s)
- David A. Guertin
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Center for Integrative Cancer Research at MIT, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Deanna M. Stevens
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
| | - Maki Saitoh
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Stephanie Kinkel
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Center for Integrative Cancer Research at MIT, 77 Massachusetts Avenue, Cambridge, MA 02139
| | | | - Joon-Ho Sheen
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Center for Integrative Cancer Research at MIT, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - David J. Mullholland
- Molecular & Medical Pharmacology, UCLA School of Medicine, 650 Charles E Young, Dr, South, CHS 23-234, Los Angeles, CA, 90095, USA
| | - Mark A. Magnuson
- Molecular Physiology an Biophysics and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Hong Wu
- Molecular & Medical Pharmacology, UCLA School of Medicine, 650 Charles E Young, Dr, South, CHS 23-234, Los Angeles, CA, 90095, USA
| | - David M. Sabatini
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Center for Integrative Cancer Research at MIT, 77 Massachusetts Avenue, Cambridge, MA 02139
- The Broad Institute, 7 Cambridge Center, Cambridge, MA 02141 USA
| |
Collapse
|
125
|
Liu P, Li S, Gan L, Kao TP, Huang H. A transcription-independent function of FOXO1 in inhibition of androgen-independent activation of the androgen receptor in prostate cancer cells. Cancer Res 2009; 68:10290-9. [PMID: 19074897 DOI: 10.1158/0008-5472.can-08-2038] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Increasing evidence suggests that aberrant activation of the androgen receptor (AR) plays a pivotal role in the development and progression of androgen depletion-independent prostate cancer (PCa) after androgen deprivation therapy. Here, we show that loss of the PTEN tumor suppressor gene is associated with hyperactivation of the AR in human PCa cell lines. This effect is mediated primarily by its downstream effector FOXO1. In addition to the inhibition of androgenic activation of the AR, forced expression of FOXO1 in PTEN-negative PCa cells also inhibits androgen-independent activation of the AR in a manner independent of FOXO1 transcriptional function. In contrast, silencing of FOXO1 in PTEN-positive cells not only increases the basal activity of the AR in the absence of androgens, it also markedly sensitizes the AR activation by low levels of androgens or nonandrogenic factors such as interleukin-6. FOXO1-mediated inhibition of the AR is partially attenuated by the histone deacetylase (HDAC) inhibitor trichostatin A. Accordingly, FOXO1 interacts with HDAC3 as shown by coimmunoprecipitation assays, and cotransfection of cells with FOXO1 and HDAC3, but not HDAC1 and HDAC2, results in a greater inhibition of AR activity than in cells transfected with FOXO1 or HDAC3 individually. Together, our findings define a novel corepressor function of FOXO1 in inhibition of androgen-independent activation of the AR.
Collapse
Affiliation(s)
- Ping Liu
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
126
|
Ma Q, Fu W, Li P, Nicosia SV, Jenster G, Zhang X, Bai W. FoxO1 mediates PTEN suppression of androgen receptor N- and C-terminal interactions and coactivator recruitment. Mol Endocrinol 2008; 23:213-25. [PMID: 19074551 DOI: 10.1210/me.2008-0147] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
FoxO (mammalian forkhead subclass O) proteins are transcription factors acting downstream of the PTEN (phosphatase and tensin homolog deleted on chromosome 10) tumor suppressor. Their activity is negatively regulated by AKT-mediated phosphorylation. Our previous studies showed that the transcriptional activity of the androgen receptor (AR) was inhibited by PTEN in an AKT-sensitive manner. Here, we report the repression of the activity of the full-length AR and its N-terminal domain by FoxO1 and the participation of FoxO1 in AR inhibition by PTEN. Ectopic expression of active FoxO1 decreased the transcriptional activity of AR as well as androgen-induced cell proliferation and production of prostate-specific antigen. FoxO1 knock down by RNA interference increased the transcriptional activity of the AR in PTEN-intact cells and relieved its inhibition by ectopic PTEN in PTEN-null cells. Mutational analysis revealed that FoxO1 fragment 150-655, which contains the forkhead box and C-terminal activation domain, was required for AR inhibition. Mammalian two-hybrid and glutathione-S-transferase pull-down assays demonstrated that the inhibition of AR activity by PTEN through FoxO1 involved the interference of androgen-induced interaction of the N- and C-termini of the AR and the recruitment of the p160 coactivators to its N terminus and to the androgen response elements of natural AR target genes. These studies reveal new mechanisms for the inhibition of AR activity by PTEN-FoxO axis and establish FoxO proteins as important nuclear factors that mediate the mutual antagonism between AR and PTEN tumor suppressor in prostate cancer cells.
Collapse
Affiliation(s)
- Qiuping Ma
- Department of Pathology, University of South Florida, College of Medicine, Tampa, Florida 33612-4799, USA
| | | | | | | | | | | | | |
Collapse
|
127
|
Song K, Wang H, Krebs TL, Kim SJ, Danielpour D. Androgenic control of transforming growth factor-beta signaling in prostate epithelial cells through transcriptional suppression of transforming growth factor-beta receptor II. Cancer Res 2008; 68:8173-82. [PMID: 18829577 DOI: 10.1158/0008-5472.can-08-2290] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The androgen receptor cross-talks with transforming growth factor-beta (TGF-beta) through mechanisms that remain poorly understood. Here we provide strong evidence that 5alpha-dihydrotestosterone (DHT) intercepts the ability of prostate epithelial cells to undergo TGF-beta-induced apoptosis, and present a new model for this androgenic effect. We report that DHT decreases the level of TGF-beta receptor II (TbetaRII) through a transcriptional mechanism, leading to suppression of the ability of TGF-beta to down-regulate expression of Bcl-xL and cyclin Ds, activate caspase-3, and induce apoptosis. Promoter analysis, DNA pulldown, and electrophoretic mobility shift assays support that transcriptional down-regulation of TbetaRII by DHT occurs through Sp1/Sp3 response elements, with the binding of Sp1 to the TbetaRII promoter being suppressed by DHT, largely driven by loss of Sp1 protein and/or activity. These results provide fresh insight on the mechanism of growth control by androgens and the progression of prostate cancer to androgen independence. [Cancer Res 2008;68(19):8173-82].
Collapse
Affiliation(s)
- Kyung Song
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
128
|
Haga K, Tomioka A, Liao CP, Kimura T, Matsumoto H, Ohno I, Hermann K, Logg CR, Jiao J, Tanaka M, Hirao Y, Wu H, Kruse CA, Roy-Burman P, Kasahara N. PTEN knockout prostate cancer as a model for experimental immunotherapy. J Urol 2008; 181:354-62. [PMID: 19010487 DOI: 10.1016/j.juro.2008.08.124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Indexed: 11/24/2022]
Abstract
PURPOSE Testing immunotherapeutic strategies for prostate cancer has been impeded by the lack of relevant tumor models in immunocompetent animals. This opportunity is now provided by the recent development of prostate specific PTEN knockout mice, which show spontaneous development of true adenocarcinoma arising from prostate epithelium and more faithfully recapitulate the human disease than any previous model. We investigated the feasibility of using tumor cells derived from this model to test tumor vaccination and adoptive immunotherapeutic strategies for prostate cancer. MATERIALS AND METHODS PTEN-CaP8 adenocarcinoma cells derived from the biallelic PTEN knockout prostate cancer model were used to vaccinate nontumor bearing litter mates. Tumor specific effector cells were generated from splenocytes of vaccinated mice by mixed lymphocyte-tumor reactions, and antiproliferative effects and cytokine generation were examined in vitro. The effect of vaccination or adoptive immunotherapy on luciferase marked PTEN-CaP8 subcutaneous tumors was monitored by tumor volumetric measurements and noninvasive bioluminescence imaging. RESULTS Vaccination of litter mate mice with irradiated PTEN-CaP8 cells showed a significant prophylactic effect against the subsequent tumor challenge. Effector cells harvested from vaccinated litter mates showed significant interferon-gamma secretion upon co-incubation with PTEN-CaP8 target cells and they were capable of efficient target cell growth inhibition in vitro. Intratumor adoptive transfer of effector cells resulted in significant growth inhibition of preestablished prostate tumors in vivo. CONCLUSIONS The PTEN knockout model serves as a highly useful model in which to investigate tumor cell vaccination and adoptive immunotherapeutic strategies in the context of true adenocarcinoma of the prostate. This model should accelerate efforts to develop effective immunotherapies for human prostate cancer.
Collapse
Affiliation(s)
- Kazunori Haga
- Department of Medicine, University of California-Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Garcia JA, Danielpour D. Mammalian target of rapamycin inhibition as a therapeutic strategy in the management of urologic malignancies. Mol Cancer Ther 2008; 7:1347-54. [PMID: 18566209 DOI: 10.1158/1535-7163.mct-07-2408] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a protein kinase that regulates protein translation, cell growth, and apoptosis. Recently, there has been an enormous increase in our understanding on molecular mechanisms underlying the therapeutics of rapamycin in cancer. Alterations in the pathway regulating mTOR occur in many solid malignancies including prostate, bladder, and kidney cancer; in vitro and in vivo models of prostate and bladder cancer have established the importance of the mTOR pathway in control of cancer progression and metastasis. Temsirolimus (Torisel) and everolimus (RAD-001), two ester analogues of rapamycin, as well as rapamycin itself have clear antitumor activity in in vitro and in vivo models and are under clinical trial investigations for prostate and bladder cancer. Phase II and III trials have already established the clinical efficacy of temsirolimus in renal cancer, and current renal trials are evaluating the combined effects of vascular endothelial growth factor and mTOR inhibition. Ongoing studies in prostate and bladder cancer will soon define the activity and safety profiles of everolimus and temsirolimus. Recent molecular advances have uncovered a startling complexity in the macromolecular function of mTOR complexes, with the identification of new mTOR partners (raptor, rictor, FKBP38, PRAS40, and mSIN1), putative cancer therapeutic/prognostic targets for future clinical trials.
Collapse
Affiliation(s)
- Jorge A Garcia
- Department of Solid Tumor Oncology and Urology, Taussig Cancer Institute, Cleveland, Ohio, USA
| | | |
Collapse
|
130
|
Hormone refractory prostate cancer: Lessons learned from the PTEN prostate cancer model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:87-95. [PMID: 18497033 DOI: 10.1007/978-0-387-69080-3_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
131
|
Correction: Role of Pten in Hormone Refractory Prostate Cancer. Cancer Res 2007. [DOI: 10.1158/0008-5472.can-67-16-cor2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|