1
|
Zhang Z, Shen Q, Ji Y, Ma Y, Hou H, Yang H, Zhu Y, Chen Y, Hu Y. Structural Optimization of Isoquinoline Derivatives from Lycobetaine and Their Inhibitory Activity against Neuroendocrine Prostate Cancer Cells. Molecules 2024; 29:4503. [PMID: 39339498 PMCID: PMC11435415 DOI: 10.3390/molecules29184503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly aggressive cancer that is resistant to hormone therapy and characterized by poor prognosis, as well as limited therapeutic options. Since the natural product lycobetaine was reported to exhibit good antitumor activities against various types of cancers, we initially simplified the scaffold of lycobetaine to obtain the active compound 1, an isoquinoline derivative with an aryl moiety substitution at the 4-position, which showed apparent antiproliferative activities against NPEC cell line LASCPC-01 in vitro. Subsequently, we carried out structural optimization and systematic structure-activity relationship (SAR) studies on compound 1, leading to the discovery of compound 46, which demonstrated potent inhibitory activities against the LASCPC-01 cell line with an IC50 value of 0.47 μM. Moreover, compound 46 displayed remarkable selectivity over prostate cancer cell line PC-3 with a selectivity index greater than 190-fold. Further cell-based mechanism studies revealed that compound 46 and lycobetaine can effectively induce G1 cell cycle arrest and apoptosis dose dependently. However, lycobetaine inhibited the expression of neuroendocrine markers, while compound 46 slightly upregulated these proteins. This suggested that compound 46 might exert its antitumor activities through a different mechanism than lycobetaine, warranting further study.
Collapse
Affiliation(s)
- Zhuo Zhang
- School of Chinese Materia Medica, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China;
| | - Qianqian Shen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.S.); (H.H.); (Y.C.)
| | - Yiyi Ji
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China;
| | - Yanjie Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China; (Y.M.); (H.Y.)
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Haiyang Hou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.S.); (H.H.); (Y.C.)
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Huajie Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China; (Y.M.); (H.Y.)
| | - Yinjie Zhu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China;
| | - Yi Chen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (Q.S.); (H.H.); (Y.C.)
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Youhong Hu
- School of Chinese Materia Medica, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China;
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China; (Y.M.); (H.Y.)
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1st Xiangshan Branch Alley, Hangzhou 310024, China
| |
Collapse
|
2
|
Lewis AR, Costello BA, Quevedo F, Pagliaro LC, Sanhueza C, Weinshilboum RM, Kalari KR, Wang L, Kohli M, Tan W, Giridhar KV. Dynamic assessment of serum chromogranin A and treatment response with abiraterone acetate in metastatic castration-resistant prostate cancer. Prostate 2023; 83:649-655. [PMID: 36924119 PMCID: PMC11537207 DOI: 10.1002/pros.24498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 12/10/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVE Elevated serum chromogranin A (CGA) is associated with intrinsic or treatment-related neuroendocrine differentiation (NED) in men with metastatic castration-resistant prostate cancer (mCRPC). Fluctuations in serum CGA during treatment of mCRPC have had conflicting results. We analyzed the impact of (i) rising serum CGA and (ii) baseline CGA/PSA ratio during treatment to identify associations with abiraterone acetate (AA) therapy. METHODS Between June 2013 and August 2015, 92 men with mCRPC were enrolled in a prospective trial with uniform serum CGA processing performed before initiating abiraterone acetate/prednisone (AA/P) and serially after 12 weeks of AA/P treatments. Serum CGA was measured using a homogenous automated immunofluorescent assay. Patients receiving proton pump inhibitors or with abnormal renal function were excluded due to possible false elevations of serum CGA (n = 21 excluded), therefore 71 patients were analyzed. All patients underwent a composite response assessment at 12-weeks. Kaplan-Meier estimates and Cox Regression models were used to calculate the association with time-to-treatment failure analyses and overall survival. RESULTS An increase in chromogranin was associated with a lower risk of treatment failure (hazard ratio [HR]: 0.52, p = 0.0181). The median CGA/PSA ratio was 7.8 (2.6-16.0) and an elevated pretreatment CGA/PSA ratio above the median was associated with a lower risk of treatment failure (HR: 0.54 p value = 0.0185). An increase in CGA was not found to be associated with OS (HR: 0.71, 95% CI: 0.42-1.21, p = 0.207). An elevated baseline CGA/PSA ratio was not associated with OS (HR: 0.62, 95% CI: 0.37-1.03, p = 0.062). An increase in PSA after 12 weeks of treatment was associated with an increased risk of treatment failure (HR: 4.14, CI: 2.21-7.73, p = < 0.0001) and worse OS (HR: 2.93, CI: 1.57-4.45, p = < 0.0001). CONCLUSIONS We show that an increasing chromogranin on AA/P and an elevated baseline CGA/PSA in patients with mCRPC were associated with a favorable response to AA/P with no changes in survival. There may be limited clinical utility in serum CGA testing to evaluate for lethal NED as AA/P did not induce lethal NED in this cohort. This highlights that not all patients with an increasing CGA have a worse OS.
Collapse
Affiliation(s)
- Akeem R Lewis
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Manish Kohli
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Winston Tan
- Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
3
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
4
|
Kannan A, Clouston D, Frydenberg M, Ilic D, Karim MN, Evans SM, Toivanen R, Risbridger GP, Taylor RA. Neuroendocrine cells in prostate cancer correlate with poor outcomes: a systematic review and meta-analysis. BJU Int 2021; 130:420-433. [PMID: 34784097 DOI: 10.1111/bju.15647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To perform a systematic review and meta-analysis of the literature to understand the variation in the reporting of neuroendocrine staining and determine the influence of reporting neuroendocrine staining at diagnosis on patient outcomes. METHODS Medical databases were searched to identify studies in which adenocarcinoma specimens were stained with any of the following four neuroendocrine markers: chromogranin A (CgA), neuron-specific enolase (NSE), synaptophysin and CD56. The prevalence of neuroendocrine staining and correlation of the prevalence of neuroendocrine staining to patient outcomes were analysed using a random-effects model. All statistical tests were two-sided. RESULTS Sixty-two studies spanning 7616 patients were analysed. The pooled prevalence for the most common marker, CgA (41%), was similar to that of NSE (39%) and higher than that of synaptophysin (31%). The prevalence of CgA staining was significantly influenced by reporting criteria, where objective thresholds reduced the variation in prevalence to 26%. No correlation was found between CgA prevalence and tumour grade. Patients positive for CgA staining using objective criteria had more rapid biochemical progression (hazard ratio [HR] 1.98, 95% confidence interval [CI] 1.49 to 2.65) and poorer prostate cancer-specific survival (HR 7.03, 95% CI 2.55 to 19.39) compared to negative patients, even among those with low-risk cancers. CONCLUSION Discrepancies in the reported prevalence of neuroendocrine cells in adenocarcinoma are driven by the inconsistent scoring criteria. This study unequivocally demonstrates that when neuroendocrine cell staining is assessed with objective criteria it identifies patients with poor clinical outcomes. Future studies are needed to determine the exact quantifiable thresholds for use in reporting neuroendocrine cell staining to identify patients at higher risk of progression.
Collapse
Affiliation(s)
- Ashwini Kannan
- Department of Anatomy and Developmental Biology and Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Vic., Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | | | - Mark Frydenberg
- Department of Anatomy and Developmental Biology and Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Vic., Australia.,Department of Surgery, Monash University, Melbourne, Vic., Australia.,Department of Urology, Cabrini Institute, Cabrini Health, Melbourne, Vic., Australia
| | - Dragan Ilic
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Md Nazmul Karim
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Sue M Evans
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic., Australia.,Victorian Cancer Registry, Cancer Council Victorian, Melbourne, Vic., Australia
| | - Roxanne Toivanen
- Department of Anatomy and Developmental Biology and Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Vic., Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology and Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Vic., Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Renea A Taylor
- Department of Anatomy and Developmental Biology and Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, Vic., Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
5
|
Bery F, Cancel M, Chantôme A, Guibon R, Bruyère F, Rozet F, Mahéo K, Fromont G. The Calcium-Sensing Receptor is A Marker and Potential Driver of Neuroendocrine Differentiation in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12040860. [PMID: 32252342 PMCID: PMC7226072 DOI: 10.3390/cancers12040860] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanisms underlying neuroendocrine (NE) differentiation in prostate cancer (PCa) remain mostly uncharacterized. Since a deregulated calcium homeostasis has been reported in neuroendocrine prostate cancer (NEPC), we explored herein the link between NE differentiation and the calcium-sensing receptor (CaSR). CaSR expression was evaluated by immunohistochemistry-together with NE markers-on tissue microarrays containing samples of normal prostate, localized PCa, metastatic castration resistant PCa (MCRPC) and NEPC. In prostate tissues, we observed a strong association between CaSR and chromogranin expression. Both markers were strongly expressed in all cases of NEPC and co-expression was confirmed by double immunostaining. In MCRPC, the expression of CaSR was significantly associated with shorter overall survival. The involvement of CaSR in NE differentiation was evaluated in PCa cell lines. Inhibition of CaSR led to decrease the expression of neuronal (NSE, βtubulinIII) and NE (chromogranin, synaptophysin) markers in the NE PCa cell line NCI-H660. A decrease of neuronal and NE markers was also observed in siCaSR-transfected PC3 and 22RV1 cells, respectively, whereas CaSR activation increased both NSE and synaptophysin expression in PC3 cells. These results strongly suggest that CaSR is a marker and a driver of NE differentiation in PCa and emphasize the potential of CaSR directed therapy for NEPC patients.
Collapse
Affiliation(s)
- Fanny Bery
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
| | - Mathilde Cancel
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
- Department of Oncology, CHRU Bretonneau, CEDEX 9, F-37044 Tours, France
| | - Aurélie Chantôme
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
| | - Roseline Guibon
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
- Department of Pathology CHRU Bretonneau, CEDEX 9, F-37044 Tours, France
| | - Franck Bruyère
- Department of Urology, CHRU Bretonneau, CEDEX 9, F-37044 Tours, France;
| | - François Rozet
- Institut Mutualiste Montsouris, Department of Urology, F-75014 Paris, France;
| | - Karine Mahéo
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
| | - Gaëlle Fromont
- Inserm N2C UMR1069 “Nutrition, Croissance et Cancer” Université de Tours, CEDEX 1, F-37032 Tours, France; (F.B.); (M.C.); (A.C.); (R.G.); (K.M.)
- Department of Pathology CHRU Bretonneau, CEDEX 9, F-37044 Tours, France
- Correspondence: ; Tel.: +33-(0)2-47-47-82-72
| |
Collapse
|
6
|
Hoang DT, Iczkowski KA, Kilari D, See W, Nevalainen MT. Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: Opportunities for therapeutic targeting from multiple angles. Oncotarget 2017; 8:3724-3745. [PMID: 27741508 PMCID: PMC5356914 DOI: 10.18632/oncotarget.12554] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/29/2016] [Indexed: 12/25/2022] Open
Abstract
Despite aggressive treatment for localized cancer, prostate cancer (PC) remains a leading cause of cancer-related death for American men due to a subset of patients progressing to lethal and incurable metastatic castrate-resistant prostate cancer (CRPC). Organ-confined PC is treated by surgery or radiation with or without androgen deprivation therapy (ADT), while options for locally advanced and disseminated PC include radiation combined with ADT, or systemic treatments including chemotherapy. Progression to CRPC results from failure of ADT, which targets the androgen receptor (AR) signaling axis and inhibits AR-driven proliferation and survival pathways. The exact mechanisms underlying the transition from androgen-dependent PC to CRPC remain incompletely understood. Reactivation of AR has been shown to occur in CRPC despite depletion of circulating androgens by ADT. At the same time, the presence of AR-negative cell populations in CRPC has also been identified. While AR signaling has been proposed as the primary driver of CRPC, AR-independent signaling pathways may represent additional mechanisms underlying CRPC progression. Identification of new therapeutic strategies to target both AR-positive and AR-negative PC cell populations and, thereby, AR-driven as well as non-AR-driven PC cell growth and survival mechanisms would provide a two-pronged approach to eliminate CRPC cells with potential for synthetic lethality. In this review, we provide an overview of AR-dependent and AR-independent molecular mechanisms which drive CRPC, with special emphasis on the role of the Jak2-Stat5a/b signaling pathway in promoting castrate-resistant growth of PC through both AR-dependent and AR-independent mechanisms.
Collapse
Affiliation(s)
- David T Hoang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William See
- Department of Urology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology/Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
7
|
Mauri G, Jachetti E, Comuzzi B, Dugo M, Arioli I, Miotti S, Sangaletti S, Di Carlo E, Tripodo C, Colombo MP. Genetic deletion of osteopontin in TRAMP mice skews prostate carcinogenesis from adenocarcinoma to aggressive human-like neuroendocrine cancers. Oncotarget 2016; 7:3905-20. [PMID: 26700622 PMCID: PMC4826179 DOI: 10.18632/oncotarget.6678] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022] Open
Abstract
Osteopontin (OPN) is a secreted glycoprotein, that belongs to the non-structural extracellular matrix (ECM), and its over expression in human prostate cancer has been associated with disease progression, androgen independence and metastatic ability. Nevertheless, the pathophysiology of OPN in prostate tumorigenesis has never been studied. We crossed TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice with OPN deficient (OPN-/-) mice and followed tumor onset and progression in these double mutants. Ultrasound examination detected the early onset of a rapidly growing, homogeneous and spherical tumor in about 60% of OPN-/- TRAMP mice. Such neoplasms seldom occurred in parental TRAMP mice otherwise prone to adenocarcinomas and were characterized for being androgen receptor negative, highly proliferative and endowed with neuroendocrine (NE) features. Gene expression profiling showed up-regulation of genes involved in tumor progression, cell cycle and neuronal differentiation in OPN-deficient versus wild type TRAMP tumors. Down-regulated genes included key genes of TGFa pathway, including SMAD3 and Filamin, which were confirmed at the protein level. Furthermore, NE genes and particularly those characterizing early prostatic lesions of OPN-deficient mice were found to correlate with those of human prostate NE tumours. These data underscore a novel role of OPN in the early stages of prostate cancer growth, protecting against the development of aggressive NE tumors.
Collapse
Affiliation(s)
- Giorgio Mauri
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Barbara Comuzzi
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Matteo Dugo
- Functional Genomics and Bioinformatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Ivano Arioli
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Silvia Miotti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Sabina Sangaletti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| | - Emma Di Carlo
- Department of Medicine and Science of Aging, Section of Anatomic Pathology and Molecular Medicine, "G. d'Annunzio" University, 66100, Chieti, Italy.,Ce.S.I. Aging Research Center, "G. d'Annunzio" University Foundation, 66100, Chieti, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90127, Palermo, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, 20133, Milano, Italy
| |
Collapse
|
8
|
Ippolito JE, Brandenburg MW, Ge X, Crowley JR, Kirmess KM, Som A, D’Avignon DA, Arbeit JM, Achilefu S, Yarasheski KE, Milbrandt J. Extracellular pH Modulates Neuroendocrine Prostate Cancer Cell Metabolism and Susceptibility to the Mitochondrial Inhibitor Niclosamide. PLoS One 2016; 11:e0159675. [PMID: 27438712 PMCID: PMC4954648 DOI: 10.1371/journal.pone.0159675] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/06/2016] [Indexed: 01/07/2023] Open
Abstract
Neuroendocrine prostate cancer is a lethal variant of prostate cancer that is associated with castrate-resistant growth, metastasis, and mortality. The tumor environment of neuroendocrine prostate cancer is heterogeneous and characterized by hypoxia, necrosis, and numerous mitoses. Although acidic extracellular pH has been implicated in aggressive cancer features including metastasis and therapeutic resistance, its role in neuroendocrine prostate cancer physiology and metabolism has not yet been explored. We used the well-characterized PNEC cell line as a model to establish the effects of extracellular pH (pH 6.5, 7.4, and 8.5) on neuroendocrine prostate cancer cell metabolism. We discovered that alkalinization of extracellular pH converted cellular metabolism to a nutrient consumption-dependent state that was susceptible to glucose deprivation, glutamine deprivation, and 2-deoxyglucose (2-DG) mediated inhibition of glycolysis. Conversely, acidic pH shifted cellular metabolism toward an oxidative phosphorylation (OXPHOS)-dependent state that was susceptible to OXPHOS inhibition. Based upon this mechanistic knowledge of pH-dependent metabolism, we identified that the FDA-approved anti-helminthic niclosamide depolarized mitochondrial potential and depleted ATP levels in PNEC cells whose effects were enhanced in acidic pH. To further establish relevance of these findings, we tested the effects of extracellular pH on susceptibility to nutrient deprivation and OXPHOS inhibition in a cohort of castrate-resistant prostate cancer cell lines C4-2B, PC-3, and PC-3M. We discovered similar pH-dependent toxicity profiles among all cell lines with these treatments. These findings underscore a potential importance to acidic extracellular pH in the modulation of cell metabolism in tumors and development of an emerging paradigm that exploits the synergy of environment and therapeutic efficacy in cancer.
Collapse
Affiliation(s)
- Joseph E. Ippolito
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| | - Matthew W. Brandenburg
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xia Ge
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jan R. Crowley
- Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kristopher M. Kirmess
- Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Avik Som
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - D. Andre D’Avignon
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, United States of America
| | - Jeffrey M. Arbeit
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kevin E. Yarasheski
- Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
9
|
Nouri M, Ratther E, Stylianou N, Nelson CC, Hollier BG, Williams ED. Androgen-targeted therapy-induced epithelial mesenchymal plasticity and neuroendocrine transdifferentiation in prostate cancer: an opportunity for intervention. Front Oncol 2014; 4:370. [PMID: 25566507 PMCID: PMC4274903 DOI: 10.3389/fonc.2014.00370] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/04/2014] [Indexed: 01/22/2023] Open
Abstract
Androgens regulate biological pathways to promote proliferation, differentiation, and survival of benign and malignant prostate tissue. Androgen receptor (AR) targeted therapies exploit this dependence and are used in advanced prostate cancer to control disease progression. Contemporary treatment regimens involve sequential use of inhibitors of androgen synthesis or AR function. Although targeting the androgen axis has clear therapeutic benefit, its effectiveness is temporary, as prostate tumor cells adapt to survive and grow. The removal of androgens (androgen deprivation) has been shown to activate both epithelial-to-mesenchymal transition (EMT) and neuroendocrine transdifferentiation (NEtD) programs. EMT has established roles in promoting biological phenotypes associated with tumor progression (migration/invasion, tumor cell survival, cancer stem cell-like properties, resistance to radiation and chemotherapy) in multiple human cancer types. NEtD in prostate cancer is associated with resistance to therapy, visceral metastasis, and aggressive disease. Thus, activation of these programs via inhibition of the androgen axis provides a mechanism by which tumor cells can adapt to promote disease recurrence and progression. Brachyury, Axl, MEK, and Aurora kinase A are molecular drivers of these programs, and inhibitors are currently in clinical trials to determine therapeutic applications. Understanding tumor cell plasticity will be important in further defining the rational use of androgen-targeted therapies clinically and provides an opportunity for intervention to prolong survival of men with metastatic prostate cancer.
Collapse
Affiliation(s)
- Mannan Nouri
- Vancouver Prostate Centre , Vancouver, BC , Canada ; The University of British Columbia , Vancouver, BC , Canada
| | - Ellca Ratther
- Australian Prostate Cancer Research Centre Queensland, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Australian Prostate Cancer Research Centre Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia
| | - Nataly Stylianou
- Australian Prostate Cancer Research Centre Queensland, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Australian Prostate Cancer Research Centre Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre Queensland, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Australian Prostate Cancer Research Centre Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia
| | - Brett G Hollier
- Australian Prostate Cancer Research Centre Queensland, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Australian Prostate Cancer Research Centre Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre Queensland, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Australian Prostate Cancer Research Centre Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland University of Technology , Brisbane, QLD , Australia ; Department of Surgery, St Vincent's Hospital, The University of Melbourne , Melbourne, VIC , Australia ; Monash University , Melbourne, VIC , Australia
| |
Collapse
|
10
|
Pernicová Z, Slabáková E, Fedr R, Šimečková Š, Jaroš J, Suchánková T, Bouchal J, Kharaishvili G, Král M, Kozubík A, Souček K. The role of high cell density in the promotion of neuroendocrine transdifferentiation of prostate cancer cells. Mol Cancer 2014; 13:113. [PMID: 24884804 PMCID: PMC4229954 DOI: 10.1186/1476-4598-13-113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 05/13/2014] [Indexed: 11/13/2022] Open
Abstract
Background Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. Results In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3′, 5′-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. Conclusions Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v,v,i, Královopolská 135, CZ-612 65 Brno, Czech Republic.
| |
Collapse
|
11
|
Terry S, Beltran H. The many faces of neuroendocrine differentiation in prostate cancer progression. Front Oncol 2014; 4:60. [PMID: 24724054 PMCID: PMC3971158 DOI: 10.3389/fonc.2014.00060] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/12/2014] [Indexed: 12/15/2022] Open
Abstract
In normal prostate, neuroendocrine (NE) cells are rare and interspersed among the epithelium. These cells are believed to provide trophic signals to epithelial cell populations through the secretion of an abundance of neuropeptides that can diffuse to influence surrounding cells. In the setting of prostate cancer (PC), NE cells can also stimulate surrounding prostate adenocarcinoma cell growth, but in some cases adenocarcinoma cells themselves acquire NE characteristics. This epithelial plasticity is associated with decreased androgen receptor (AR) signaling and the accumulation of neuronal and stem cell characteristics. Transformation to an NE phenotype is one proposed mechanism of resistance to contemporary AR-targeted treatments, is associated with poor prognosis, and thought to represent up to 25% of lethal PCs. Importantly, the advent of high-throughput technologies has started to provide clues for understanding the complex molecular profiles of tumors exhibiting NE differentiation. Here, we discuss these recent advances, the multifaceted manner by which an NE-like state may arise during the different stages of disease progression, and the potential benefit of this knowledge for the management of patients with advanced PC.
Collapse
Affiliation(s)
- Stéphane Terry
- U955, Institut Mondor de Recherche Biomédicale, INSERM , Créteil , France ; UMR 3244, Institut Curie , Paris , France
| | - Himisha Beltran
- Division of Hematology and Medical Oncology, Weill Cornell Medical College , New York, NY , USA
| |
Collapse
|
12
|
Ippolito JE, Piwnica-Worms D. A fluorescence-coupled assay for gamma aminobutyric acid (GABA) reveals metabolic stress-induced modulation of GABA content in neuroendocrine cancer. PLoS One 2014; 9:e88667. [PMID: 24551133 PMCID: PMC3923810 DOI: 10.1371/journal.pone.0088667] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 01/15/2014] [Indexed: 11/18/2022] Open
Abstract
Pathways involved in the synthesis of the neurotransmitter gamma-aminobutyric acid (GABA) have been implicated in the pathogenesis of high grade neuroendocrine (NE) neoplasms as well as neoplasms from a non-NE lineage. Using The Cancer Genome Atlas, overexpression of the GABA synthetic enzyme, glutamate decarboxylase 1 (GAD1), was found to be associated with decreased disease free-survival in prostate adenocarcinoma and decreased overall survival in clear cell renal cell carcinomas. Furthermore, GAD1 was found to be expressed in castrate-resistant prostate cancer cell lines, but not androgen-responsive cell lines. Using a novel fluorescence-coupled enzymatic microplate assay for GABA mediated through reduction of resazurin in a prostate neuroendocrine carcinoma (PNEC) cell line, acid microenvironment-induced stress increased GABA levels while alkaline microenvironment-induced stress decreased GABA through modulation of GAD1 and glutamine synthetase (GLUL) activities. Moreover, glutamine but not glucose deprivation decreased GABA through modulation of GLUL. Consistent with evidence in prokaryotic and eukaryotic organisms that GABA synthesis mediated through GAD1 may play a crucial role in surviving stress, GABA may be an important mediator of stress survival in neoplasms. These findings identify GABA synthesis and metabolism as a potentially important pathway for regulating cancer cell stress response as well as a potential target for therapeutic strategies.
Collapse
Affiliation(s)
- Joseph E. Ippolito
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (JEI); (DP-W)
| | - David Piwnica-Worms
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- BRIGHT Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cancer Systems Imaging, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (JEI); (DP-W)
| |
Collapse
|
13
|
Cross modulation between the androgen receptor axis and protocadherin-PC in mediating neuroendocrine transdifferentiation and therapeutic resistance of prostate cancer. Neoplasia 2014; 15:761-72. [PMID: 23814488 DOI: 10.1593/neo.122070] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/14/2013] [Accepted: 04/15/2013] [Indexed: 12/31/2022] Open
Abstract
Castration-resistant prostate cancers (CRPCs) that relapse after androgen deprivation therapies (ADTs) are responsible for the majority of mortalities from prostate cancer (PCa). While mechanisms enabling recurrent activity of androgen receptor (AR) are certainly involved in the development of CRPC, there may be factors that contribute to the process including acquired neuroendocrine (NE) cell-like behaviors working through alternate (non-AR) cell signaling systems or AR-dependent mechanisms. In this study, we explore the potential relationship between the AR axis and a novel putative marker of NE differentiation, the human male protocadherin-PC (PCDH-PC), in vitro and in human situations. We found evidence for an NE transdifferentiation process and PCDH-PC expression as an early-onset adaptive mechanism following ADT and elucidate AR as a key regulator of PCDH-PC expression. PCDH-PC overexpression, in turn, attenuates the ligand-dependent activity of the AR, enabling certain prostate tumor clones to assume a more NE phenotype and promoting their survival under diverse stress conditions. Acquisition of an NE phenotype by PCa cells positively correlated with resistance to cytotoxic agents including docetaxel, a taxane chemotherapy approved for the treatment of patients with metastatic CRPC. Furthermore, knockdown of PCDH-PC in cells that have undergone an NE transdifferentiation partially sensitized cells to docetaxel. Together, these results reveal a reciprocal regulation between the AR axis and PCDH-PC signals, observed both in vitro and in vivo, with potential implications in coordinating NE transdifferentiation processes and progression of PCa toward hormonal and chemoresistance.
Collapse
|
14
|
Geomela PA, Kontos CK, Yiotakis I, Fragoulis EG, Scorilas A. L-DOPA decarboxylase mRNA expression is associated with tumor stage and size in head and neck squamous cell carcinoma: a retrospective cohort study. BMC Cancer 2012; 12:484. [PMID: 23083099 PMCID: PMC3495033 DOI: 10.1186/1471-2407-12-484] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/17/2012] [Indexed: 12/22/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) represents one of the most commonly diagnosed malignancies worldwide. The DDC gene encodes L-DOPA decarboxylase, an enzyme catalyzing the decarboxylation of L-DOPA to dopamine. We have recently shown that DDC mRNA is a significant predictor of patients’ prognosis in colorectal adenocarcinoma and prostate cancer. The aim of the current study was to analyze the DDC mRNA expression in HNSCC patients. Methods 53 malignant tumors were resected from the larynx, pharynx, tongue, buccal mucosa, parotid glands, and nasal cavity, as well as from 34 adjacent non-cancerous tissues of HNSCC patients, and were homogenized. Total RNA was isolated and converted into first-strand cDNA. An ultrasensitive real-time PCR method based on the SYBR Green chemistry was used for DDC mRNA quantification in head and neck tissue specimens. Relative quantification was performed using the comparative Ct (2-ddCt) method. Results DDC mRNA levels were lower in squamous cell carcinomas (SCCs) of the larynx and tongue than in adjacent non-cancerous tissue specimens. Furthermore, low DDC mRNA expression was noticed in laryngeal and tongue tumors of advanced TNM stage or bigger size, compared to early-stage or smaller tumors, respectively. No statistically significant differences were observed between SCCs resected from pharynx, buccal mucosa, or nasal cavity, and their normal counterparts. Conclusion This is the first study examining the DDC mRNA expression in HNSCC. According to our results, DDC mRNA expression may constitute a potential prognostic biomarker in tongue and/or larynx SCCs, which principally represent the overwhelming majority of HNSCC cases.
Collapse
Affiliation(s)
- Panagiota-Aikaterini Geomela
- Department of Biochemistry and Molecular Biology, University of Athens, Panepistimiopolis, Athens 15701, Greece.
| | | | | | | | | |
Collapse
|
15
|
Thomas C, Wafa LA, Lamoureux F, Cheng H, Fazli L, Gleave ME, Rennie PS. Carbidopa enhances antitumoral activity of bicalutamide on the androgen receptor-axis in castration-resistant prostate tumors. Prostate 2012; 72:875-85. [PMID: 22072572 DOI: 10.1002/pros.21490] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Accepted: 08/24/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Response to bicalutamide after castration failure is not durable and treatment options at this stage are limited. Carbidopa, an L-dopa decarboxylase (AR-coactivator) inhibitor, has been shown to retard prostate tumor growth/PSA production in xenografts. Here, we hypothesize that pharmacological targeting of the AR-axis by combination treatment with bicalutamide plus carbidopa significantly enhances antitumoral activity in vitro and in vivo compared to monotherapy with either drug. METHODS Carbidopa was tested for its ability to enhance the effects of bicalutamide on cell viability, apoptosis and PSA transactivation in LNCaP and C4-2 cells. The castration-resistant prostate cancer (CRPC) LNCaP xenograft tumor model was used in vivo. After CRPC progression, mice were treated with carbidopa (50 mg/kg) and bicalutamide (50 mg/kg) as monotherapy or in combination. Tumor volume and serum PSA were evaluated weekly. RESULTS Combination treatment of carbidopa plus bicalutamide significantly inhibited cell viability in both cell lines and induced apoptosis. The combination treatment also decreased androgen-induced PSA transactivation by 62.6% in LNCaP cells and by 55.6% in C4-2 cells compared to control, while bicalutamide monotherapy reduced PSA levels by 27.5% and 29.1% in LNCaP and C4-2 cells. In vivo, bicalutamide monotherapy delayed LNCaP CRPC tumor growth rate by 72.2%, while combination treatment reduced tumor growth by 84.4% compared to control. Serum PSA was also reduced 70.6% with bicalutamide monotherapy, while combination therapy reduced PSA levels by 76.7% compared to control. CONCLUSIONS This study demonstrates preclinical proof-of-principle that pharmacological targeting of prostate tumors by combination treatment of bicalutamide plus carbidopa significantly reduces AR activity, and thereby delays CRPC tumor progression in vivo.
Collapse
Affiliation(s)
- Christian Thomas
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | | | | | | | | | | | | |
Collapse
|
16
|
Koutalellis G, Stravodimos K, Avgeris M, Mavridis K, Scorilas A, Lazaris A, Constantinides C. L-dopa decarboxylase (DDC) gene expression is related to outcome in patients with prostate cancer. BJU Int 2012; 110:E267-73. [PMID: 22571720 DOI: 10.1111/j.1464-410x.2012.11152.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
UNLABELLED What's known on the subject? and What does the study add? L-dopa decarboxylase (DDC) has been documented as a novel co-activator of androgen receptor transcriptional activity. Recently, it was shown that DDC gene expression is significantly higher in patients with PCa than in those with BPH. In the present study, there was a significant association between the DDC gene expression levels and the pathological stage and Gleason score of patients with prostate cancer (PCa). Moreover, DDC expression was shown to be an unfavourable prognostic marker of biochemical recurrence and disease-free survival in patients with PCa treated by radical prostatectomy. OBJECTIVE To determine whether L-dopa decarboxylase gene (DDC) expression levels in patients with prostate cancer (PCa) correlate to biochemical recurrence and disease prognosis after radical prostatectomy (RP). PATIENTS AND METHODS The present study consisted of 56 samples with confirmed malignancy from patients with PCa who had undergone RP at a single tertiary academic centre. Total RNA was isolated from tissue specimens and a SYBR Green fluorescence-based quantitative real-time polymerase chain reaction methodology was developed for the determination of DDC mRNA expression levels of the tested tissues. Follow-up time ranged between 1.0 and 62.0 months (mean ± SE, 28.6 ± 2.1 month; median, 31.5 months). Time to biochemical recurrence was defined as the interval between the surgery and the measurement of two consecutive values of prostate-specific antigen (PSA) ≥0.2 ng/mL. RESULTS DDC expression levels were found to be positively correlated with the tumour-node-metastasis stage (P = 0.021) and Gleason score (P = 0.036) of the patients with PCa. Patients with PCa with raised DDC expression levels run a significantly higher risk of biochemical recurrence after RP, as indicated by Cox proportional regression analysis (P = 0.021). Multivariate Cox proportional regression models revealed the preoperative PSA-, age- and digital rectal examination-independent prognostic value of DDC expression for the prediction of disease-free survival (DFS) among patients with PCa (P = 0.036). Kaplan-Meier survival analysis confirms the significantly shorter DFS after RP of PCa with higher DDC expression levels (P = 0.015). CONCLUSIONS This is the first study indicating the potential of DDC expression as a novel prognostic biomarker in patients with PCa who have undergone RP. For further evaluation and clinical application of the findings of the present study, a direct analysis of mRNA and/or its protein expression level in preoperative biopsy, blood serum and urine should be conducted.
Collapse
Affiliation(s)
- Georgios Koutalellis
- 1st Department of Urology, Laiko General Hospital, University of Athens, School of Medicine, Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
17
|
Wafa LA, Cheng H, Plaa N, Ghaidi F, Fukumoto T, Fazli L, Gleave ME, Cox ME, Rennie PS. Carbidopa abrogates L-dopa decarboxylase coactivation of the androgen receptor and delays prostate tumor progression. Int J Cancer 2011; 130:2835-44. [PMID: 21780103 DOI: 10.1002/ijc.26287] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 06/28/2011] [Indexed: 01/24/2023]
Abstract
The androgen receptor (AR) plays a central role in prostate cancer progression to the castration-resistant (CR) lethal state. L-Dopa decarboxylase (DDC) is an AR coactivator that increases in expression with disease progression and is coexpressed with the receptor in prostate adenocarcinoma cells, where it may enhance AR activity. Here, we hypothesize that the DDC enzymatic inhibitor, carbidopa, can suppress DDC-coactivation of AR and retard prostate tumor growth. Treating LNCaP prostate cancer cells with carbidopa in transcriptional assays suppressed the enhanced AR transactivation seen with DDC overexpression and decreased prostate-specific antigen (PSA) mRNA levels. Carbidopa dose-dependently inhibited cell growth and decreased survival in LNCaP cell proliferation and apoptosis assays. The inhibitory effect of carbidopa on DDC-coactivation of AR and cell growth/survival was also observed in PC3 prostate cancer cells (stably expressing AR). In vivo studies demonstrated that serum PSA velocity and tumor growth rates elevated ∼2-fold in LNCaP xenografts, inducibly overexpressing DDC, were reverted to control levels with carbidopa administration. In castrated mice, treating LNCaP tumors, expressing endogenous DDC, with carbidopa delayed progression to the CR state from 6 to 10 weeks, while serum PSA and tumor growth decreased 4.3-fold and 5.4-fold, respectively. Our study is a first time demonstration that carbidopa can abrogate DDC-coactivation of AR in prostate cancer cells and tumors, decrease serum PSA, reduce tumor growth and delay CR progression. Since carbidopa is clinically approved, it may be readily used as a novel therapeutic strategy to suppress aberrant AR activity and delay prostate cancer progression.
Collapse
Affiliation(s)
- Latif A Wafa
- Department of Urologic Sciences, The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Quantitative expression analysis and prognostic significance of L-DOPA decarboxylase in colorectal adenocarcinoma. Br J Cancer 2010; 102:1384-90. [PMID: 20424616 PMCID: PMC2865762 DOI: 10.1038/sj.bjc.6605654] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND L-DOPA decarboxylase (DDC) is an enzyme that catalyses, mainly, the decarboxylation of L-DOPA to dopamine and was found to be involved in many malignancies. The aim of this study was to investigate the mRNA expression levels of the DDC gene and to evaluate its clinical utility in tissues with colorectal adenocarcinoma. METHODS Total RNA was isolated from colorectal adenocarcinoma tissues of 95 patients. After having tested RNA quality, we prepared cDNA by reverse transcription. Highly sensitive quantitative real-time PCR method for DDC mRNA quantification was developed using the SYBR Green chemistry. GAPDH served as a housekeeping gene. Relative quantification analysis was performed using the comparative C(T) method (2(-DeltaDeltaC(T))). RESULTS DDC mRNA expression varied remarkably among colorectal tumours examined in this study. High DDC mRNA expression levels were found in well-differentiated and Dukes' stage A and B tumours. Kaplan-Meier survival curves showed that patients with DDC-positive tumours have significantly longer disease-free survival (P=0.009) and overall survival (P=0.027). In Cox regression analysis of the entire cohort of patients, negative DDC proved to be a significant predictor of reduced disease-free (P=0.021) and overall survival (P=0.047). CONCLUSIONS The results of the study suggest that DDC mRNA expression may be regarded as a novel potential tissue biomarker in colorectal adenocarcinoma.
Collapse
|
19
|
Tavassoli P, Wafa LA, Cheng H, Zoubeidi A, Fazli L, Gleave M, Snoek R, Rennie PS. TAF1 differentially enhances androgen receptor transcriptional activity via its N-terminal kinase and ubiquitin-activating and -conjugating domains. Mol Endocrinol 2010; 24:696-708. [PMID: 20181722 DOI: 10.1210/me.2009-0229] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Aberrant expression of androgen receptor (AR) coregulators has been linked to progression of prostate cancers to castration resistance. Using the repressed transactivator yeast two-hybrid system, we found that TATA binding protein-associated factor 1 (TAF1) interacted with the AR. In tissue microarrays, TAF1 was shown to steadily increase with duration of neoadjuvant androgen withdrawal and with progression to castration resistance. Glutathione S-transferase pulldown assays established that TAF1 bound through its acetylation and ubiquitin-activating/conjugating domains (E1/E2) directly to the AR N terminus. Coimmunoprecipitation and ChIP assays revealed colocalization of TAF1 and AR on the prostate-specific antigen promoter/enhancer in prostate cancer cells. With respect to modulation of AR activity, overexpression of TAF1 enhanced AR activity severalfold, whereas small interfering RNA knockdown of TAF1 significantly decreased AR transactivation. Although full-length TAF1 showed enhancement of both AR and some generic gene transcriptional activity, selective AR coactivator activity by TAF1 was demonstrated in transactivation experiments using cloned N-terminal kinase and E1/E2 functional domains. In keeping with AR coactivation by the ubiquitin-activating and -conjugating domain, TAF1 was found to greatly increase the cellular amount of polyubiquitinated AR. In conclusion, our results indicate that increased TAF1 expression is associated with progression of human prostate cancers to the lethal castration-resistant state. Because TAF1 is a coactivator of AR that binds and enhances AR transcriptional activity, its overexpression could be part of a compensatory mechanism adapted by cancer cells to overcome reduced levels of circulating androgens.
Collapse
Affiliation(s)
- Peyman Tavassoli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang J, Kim J, Roh M, Franco OE, Hayward SW, Wills ML, Abdulkadir SA. Pim1 kinase synergizes with c-MYC to induce advanced prostate carcinoma. Oncogene 2010; 29:2477-87. [PMID: 20140016 PMCID: PMC2861731 DOI: 10.1038/onc.2010.10] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The oncogenic PIM1 kinase has been implicated as a cofactor for c-MYC in prostate carcinogenesis. In this study, we show that in human prostate tumors, coexpression of c-MYC and PIM1 is associated with higher Gleason grades. Using a tissue recombination model coupled with lentiviral-mediated gene transfer we find that Pim1 is weakly oncogenic in naive adult mouse prostatic epithelium. However, it cooperates dramatically with c-MYC to induce prostate cancer within 6-weeks. Importantly, c-MYC/Pim1 synergy is critically dependent on Pim1 kinase activity. c-MYC/Pim1 tumors showed increased levels of the active serine-62 (S62) phosphorylated form of c-MYC. Grafts expressing a phosphomimetic c-MYCS62D mutant had higher rates of proliferation than grafts expressing wild type c-MYC but did not form tumors like c-MYC/Pim1 grafts, indicating that Pim1 cooperativity with c-MYC in vivo involves additional mechanisms other than enhancement of c-MYC activity by S62 phosphorylation. c-MYC/Pim1-induced prostate carcinomas show evidence of neuroendocrine (NE) differentiation. Additional studies, including the identification of tumor cells coexpressing androgen receptor and NE cell markers synaptophysin and Ascl1 suggested that NE tumors arose from adenocarcinoma cells through transdifferentiation. These results directly show functional cooperativity between c-MYC and PIM1 in prostate tumorigenesis in vivo and support efforts for targeting PIM1 in prostate cancer.
Collapse
Affiliation(s)
- J Wang
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Shaw G, Oliver R. Intermittent hormone therapy and its place in the contemporary endocrine treatment of prostate cancer. Surg Oncol 2009; 18:275-82. [DOI: 10.1016/j.suronc.2009.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Zhang KX, Moussavi M, Kim C, Chow E, Chen IS, Fazli L, Jia W, Rennie PS. Lentiviruses with trastuzumab bound to their envelopes can target and kill prostate cancer cells. Cancer Gene Ther 2009; 16:820-31. [PMID: 19373278 DOI: 10.1038/cgt.2009.28] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In this study, we took advantage of the overexpression of human epidermal growth factor receptor 2 (HER-2) in prostate cancers to design lentiviruses with modified envelope proteins that bind antibodies to specific cell-surface antigens. When bound to trastuzumab (Herceptin, Genentech, CA), lentiviruses were able to selectively infect androgen-sensitive LNCaP and castration-resistant C4-2 human prostate cancer cell lines, both of which express high levels of HER-2. To test for a therapeutic effect, we engineered our antibody-binding lentiviruses to express thymidine kinase, which can convert the non-toxic pro-drug ganciclovir (GCV) into a cytotoxic form. LNCaP and C4-2 cells infected by these viruses were sensitive to GCV killing. In vivo, C4-2 xenograft tumors treated either intratumorally or i.v. with trastuzumab-bound lentivirus expressed luciferase, although the latter route was less tumor specific. When a prostate-specific promoter for governing luciferase expression was combined with trastuzumab-mediated delivery, there was a further enrichment in targeting viral gene expression in prostate tumors. In conclusion, we found that although prostate cancers that express high levels of HER-2 are resistant to the killing effects of trastuzumab, they can be targeted for selective gene expression and destruction by viruses with envelope proteins engineered to bind this antibody.
Collapse
Affiliation(s)
- K-x Zhang
- Prostate Centre at Vancouver General Hospital, Vancouver, British Columbia, Canada V6H 3Z6
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Snoek R, Cheng H, Margiotti K, Wafa LA, Wong CA, Wong EC, Fazli L, Nelson CC, Gleave ME, Rennie PS. In vivo Knockdown of the Androgen Receptor Results in Growth Inhibition and Regression of Well-Established, Castration-Resistant Prostate Tumors. Clin Cancer Res 2008; 15:39-47. [DOI: 10.1158/1078-0432.ccr-08-1726] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Abstract
Neuroendocrine tumours (NETs) comprise a heterogenous group of malignancies with an often unpredictable course, and with limited treatment options. Thus, new diagnostic, prognostic, and therapeutic markers are needed. To shed new lights into the biology of NETs, we have by cDNA transcript profiling, sought to identify genes that are either up- or downregulated in NE as compared with non-NE tumour cells. A panel of six NET and four non-NET cell lines were examined, and out of 12 743 genes examined, we studied in detail the 200 most significantly differentially expressed genes in the comparison. In addition to potential new diagnostic markers (NEFM, CLDN4, PEROX2), the results point to genes that may be involved in the tumorigenesis (BEX1, TMEPAI, FOSL1, RAB32), and in the processes of invasion, progression and metastasis (MME, STAT3, DCBLD2) of NETs. Verification by real time qRT–PCR showed a high degree of consistency to the microarray results. Furthermore, the protein expression of some of the genes were examined. The results of our study has opened a window to new areas of research, by uncovering new candidate genes and proteins to be further investigated in the search for new prognostic, predictive, and therapeutic markers in NETs.
Collapse
|
25
|
Expression analysis and clinical utility of L-Dopa decarboxylase (DDC) in prostate cancer. Clin Biochem 2008; 41:1140-9. [PMID: 18586020 DOI: 10.1016/j.clinbiochem.2008.04.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Revised: 04/20/2008] [Accepted: 04/24/2008] [Indexed: 11/21/2022]
Abstract
BACKGROUND L-Dopa decarboxylase (DDC) is a pyridoxal 5'-phosphate-dependent enzyme that was found to be involved in many malignancies. The aim of this study was to investigate the mRNA expression levels of DDC in prostate tissues and to evaluate its clinical utility in prostate cancer (CaP). METHODS Total RNA was isolated from 118 tissue specimens from benign prostate hyperplasia (BPH) and CaP patients and a highly sensitive quantitative real-time RT-PCR (qRT-PCR) method for DDC mRNA quantification has been developed using the SYBR Green chemistry. LNCaP prostate cancer cell line was used as a calibrator and GAPDH as a housekeeping gene. RESULTS DDC was found to be overexpressed, at the mRNA level, in the specimens from prostate cancer patients, in comparison to those from benign prostate hyperplasia patients (p<0.001). Logistic regression and ROC analysis have demonstrated that the DDC expression has significant discriminatory value between CaP and BPH (p<0.001). DDC expression status was compared with other established prognostic factors, in prostate cancer. High expression levels of DDC were found more frequently in high Gleason's score tumors (p=0.022) as well as in advanced stage patients (p=0.032). CONCLUSIONS Our data reveal the potential of DDC expression, at the mRNA level, as a novel biomarker in prostate cancer.
Collapse
|
26
|
Bibliography. Current world literature. Adrenal cortex. Curr Opin Endocrinol Diabetes Obes 2008; 15:284-299. [PMID: 18438178 DOI: 10.1097/med.0b013e3283040e80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
D'Antonio JM, Ma C, Monzon FA, Pflug BR. Longitudinal analysis of androgen deprivation of prostate cancer cells identifies pathways to androgen independence. Prostate 2008; 68:698-714. [PMID: 18302219 DOI: 10.1002/pros.20677] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Following androgen ablation therapy, the majority of prostate cancer patients develop treatment resistance with a median time of 18-24 months to disease progression. METHODS To identify molecular targets that promote prostate cancer cell survival and contribute to androgen independence, we evaluated changes in LNCaP cell gene expression during 12 months of androgen deprivation. At time points reflecting critical growth and phenotypic changes, we performed Affymetrix expression array analysis to examine the effects of androgen deprivation during the acute response, during the period of apparent quiescence, and following the emergence of a highly proliferative, androgen-independent prostate cancer cell phenotype (LNCaP-AI). RESULTS We discovered alterations in gene expression for molecules associated with promoting prostate cancer cell growth and survival, and regulating cell cycle progression and apoptosis. Additionally, expression of AR co-regulators, adrenal androgen metabolizing enzymes, and markers of neuroendocrine disease were significantly altered. CONCLUSIONS These findings contribute greatly to our understanding of androgen-independent prostate cancer. The value of this longitudinal approach lies in the ability to examine gene expression changes throughout the adaptive response to androgen deprivation; it provides a more dynamic illustration of genes which contribute to disease progression in addition to specific genes which constitute an androgen-independent phenotype.
Collapse
Affiliation(s)
- Jason M D'Antonio
- Program in Cellular and Molecular Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | |
Collapse
|
28
|
Palmer J, Venkateswaran V, Fleshner NE, Klotz LH, Cox ME. The impact of diet and micronutrient supplements on the expression of neuroendocrine markers in murine Lady transgenic prostate. Prostate 2008; 68:345-53. [PMID: 18188867 DOI: 10.1002/pros.20692] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Neuroendocrine (NE) differentiation (NED) in prostate cancer (PCa) is associated with morbidity and death; however, the underlying cause(s) promoting NED in PCa have yet to be determined. In this study, we examined the effect of both diet and micronutrient supplementation on the expression of NE markers using the Lady (12T-10) transgenic model of PCa. Lady (12T-10) transgenic animals develop advanced adenocarcinoma with NE characteristics that exhibits metastases in approximately 80% of cases. In this model a high fat diet has been shown to increase the severity of disease, while the use of micronutrients can inhibit this progression. METHODS In this study we used immunohistochemical analysis to determine expression of the NE markers: chromogranin A (CgA), neuron-specific enolase (NSE), bombesin, parathyroid hormone-related peptide (PTHrP), neurotensin and serotonin in prostates of PCa-bearing Lady (12T-10) mice. RESULTS High fat diet was correlated with significantly elevated expression of CgA and serotonin in prostate tissue of Lady (12T-10) mice. Addition of micronutrients to the control and high fat diet reproducibly elevated PTHrP and bombesin expression and suppressed NSE expression, while prostate tissue from the control diet supplemented with micronutrients exhibited significantly lower numbers of calcitonin- and neurotensin-positive cells. CONCLUSIONS These results highlight the importance of dietary control in management of disease and identify differential changes in NE marker expression, which may be diagnostically viable in monitoring the impact of therapies on disease status.
Collapse
Affiliation(s)
- Jodie Palmer
- The Prostate Centre at Vancouver General Hospital, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
29
|
Sion-Vardy N, Priel-Cohen Z, Mermershtain W, Neulander E, Benharroch D. Ethnicity and its significance in the pathobiology of prostatic carcinoma in Southern Israel. Urol Oncol 2008; 26:31-6. [PMID: 18190827 DOI: 10.1016/j.urolonc.2007.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Revised: 01/17/2007] [Accepted: 01/18/2007] [Indexed: 11/17/2022]
Abstract
BACKGROUND Several studies have noted ethnic differences in the natural history of prostatic carcinoma. Southern Israel has been regarded as a melting pot and, perhaps more than the rest of the country, has encouraged the ingathering of immigrants from several countries, as well as a large Bedouin community. OBJECTIVES In an attempt to determine any differences that may exist in population groups in Israel, we have examined clinical and biologic markers in patients diagnosed with prostatic cancer in Southern Israel in 1996-2000. We wanted to demonstrate differences in the incidence and features of prostate carcinoma among the population groups in Southern Israel, and to evaluate their possible biologic significance. METHODS Clinical parameter features, including the ethnicity origin of patients with prostatic adenocarcinoma, were reviewed in a cohort of 189 patients seen between 1996 and 2000. Tissue sections from specimens in a subset of 40 of these patients who had undergone prostatectomy were studied by immunohistochemistry for TP53, Bcl-2, and chromogranin A using the ABC peroxidase method. These markers were chosen because of their suggested impact on the biology of this tumor. Clinical correlations were examined. RESULTS We confirm the presence of ethnic differences in the features of prostatic adenocarcinoma in our geographic area. Notably, patients of North African origin were treated surgically at a younger age than immigrants from East Europe. Higher total prostate-specific antigen levels and more robust tumor cell Bcl-2 expression were detected in the East European patients. The number of Bedouin subjects in our cohort of patients with prostatic cancer was much more limited than expected. No immigrants from Ethiopia were included in our study diagnosed with prostate carcinoma during this period. CONCLUSIONS The proportion of patients of European, especially East European, origin was relatively high among the cohort of 189. Their older age and the lower proportion of subjects that underwent surgery, together with the tendency toward higher total prostate-specific antigen levels and higher Bcl-2 expression, suggest that this ethnic group may not differ significantly from the African-American group in the United States. The low representation of Bedouin and absence of Ethiopian immigrants among our patients with prostate cancer may point to a genuinely low incidence or it may be related to inadequate medical supervision in these population groups.
Collapse
Affiliation(s)
- Netta Sion-Vardy
- Institute of Pathology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
30
|
Margiotti K, Wafa LA, Cheng H, Novelli G, Nelson CC, Rennie PS. Androgen-regulated genes differentially modulated by the androgen receptor coactivator L-dopa decarboxylase in human prostate cancer cells. Mol Cancer 2007; 6:38. [PMID: 17553164 PMCID: PMC1904238 DOI: 10.1186/1476-4598-6-38] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 06/06/2007] [Indexed: 12/22/2022] Open
Abstract
Background The androgen receptor is a ligand-induced transcriptional factor, which plays an important role in normal development of the prostate as well as in the progression of prostate cancer to a hormone refractory state. We previously reported the identification of a novel AR coactivator protein, L-dopa decarboxylase (DDC), which can act at the cytoplasmic level to enhance AR activity. We have also shown that DDC is a neuroendocrine (NE) marker of prostate cancer and that its expression is increased after hormone-ablation therapy and progression to androgen independence. In the present study, we generated tetracycline-inducible LNCaP-DDC prostate cancer stable cells to identify DDC downstream target genes by oligonucleotide microarray analysis. Results Comparison of induced DDC overexpressing cells versus non-induced control cell lines revealed a number of changes in the expression of androgen-regulated transcripts encoding proteins with a variety of molecular functions, including signal transduction, binding and catalytic activities. There were a total of 35 differentially expressed genes, 25 up-regulated and 10 down-regulated, in the DDC overexpressing cell line. In particular, we found a well-known androgen induced gene, TMEPAI, which wasup-regulated in DDC overexpressing cells, supporting its known co-activation function. In addition, DDC also further augmented the transcriptional repression function of AR for a subset of androgen-repressed genes. Changes in cellular gene transcription detected by microarray analysis were confirmed for selected genes by quantitative real-time RT-PCR. Conclusion Taken together, our results provide evidence for linking DDC action with AR signaling, which may be important for orchestrating molecular changes responsible for prostate cancer progression.
Collapse
MESH Headings
- Adenocarcinoma/enzymology
- Adenocarcinoma/metabolism
- Androgens
- Blotting, Western
- Cell Line, Tumor/metabolism
- Dopa Decarboxylase/genetics
- Dopa Decarboxylase/physiology
- Enzyme Induction/drug effects
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genetic Vectors/pharmacology
- Humans
- Male
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neoplasms, Hormone-Dependent/enzymology
- Neoplasms, Hormone-Dependent/metabolism
- Oligonucleotide Array Sequence Analysis
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Receptors, Androgen/physiology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Tetracycline/pharmacology
- Transfection
Collapse
Affiliation(s)
- Katia Margiotti
- The Prostate Centre at Vancouver General Hospital, 2660 Oak Street, V6H 3Z6, Vancouver, BC, Canada
- Department of Biopathology and Diagnostic Imaging, Tor Vergata University of Rome, Viale Oxford, 81-00133, Rome, Italy
| | - Latif A Wafa
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- The Prostate Centre at Vancouver General Hospital, 2660 Oak Street, V6H 3Z6, Vancouver, BC, Canada
| | - Helen Cheng
- The Prostate Centre at Vancouver General Hospital, 2660 Oak Street, V6H 3Z6, Vancouver, BC, Canada
| | - Giuseppe Novelli
- Department of Biopathology and Diagnostic Imaging, Tor Vergata University of Rome, Viale Oxford, 81-00133, Rome, Italy
| | - Colleen C Nelson
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- The Prostate Centre at Vancouver General Hospital, 2660 Oak Street, V6H 3Z6, Vancouver, BC, Canada
| | - Paul S Rennie
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- The Prostate Centre at Vancouver General Hospital, 2660 Oak Street, V6H 3Z6, Vancouver, BC, Canada
| |
Collapse
|