51
|
Jiang L, Shan J, Shen J, Wang Y, Yan P, Liu L, Zhao W, Xu Y, Zhu W, Su L, Chen J, Cheng F, Yao H, Xu H, Qian C, Liang Z. Androgen/androgen receptor axis maintains and promotes cancer cell stemness through direct activation of Nanog transcription in hepatocellular carcinoma. Oncotarget 2018; 7:36814-36828. [PMID: 27167111 PMCID: PMC5095041 DOI: 10.18632/oncotarget.9192] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/16/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and malignant cancers. The HCC incidence gets a strong sexual dimorphism as men are the major sufferers in this disaster. Although several studies have uncovered the presentative correlation between the axis of androgen/androgen receptor (AR) and HCC incidence, the mechanism is still largely unknown. Cancer stem cells (CSCs) are a small subgroup of cancer cells contributing to multiple tumors malignant behaviors, which play an important role in oncogenesis of various cancers including HCC. However, whether androgen/AR axis involves in regulation of HCC cells stemness remains unclear. Our previous study had identified that the pluripotency factor Nanog is not only a stemness biomarker, but also a potent regulator of CSCs in HCC. In this study, we revealed androgen/AR axis can promote HCC cells stemness by transcriptional activation of Nanog expression through directly binding to its promoter. In HCC tissues, we found that AR expression was abnormal high and got correlation with Nanog. Then, by labeling cellular endogenous Nanog with green fluorescent protein (GFP) through CRISPR/Cas9 system, it verified the co-localization of AR and Nanog in HCC cells. With in vitro experiments, we demonstrated the axis can promote HCC cells stemness, which effect is in a Nanog-dependent manner and through activating its transcription. And the xenografted tumor experiments confirmed the axis effect on tumorigenesis facilitation in vivo. Above all, we revealed a new sight of androgen/AR axis roles in HCC and provided a potential way for suppressing the axis in HCC therapy.
Collapse
Affiliation(s)
- Lupin Jiang
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Juanjuan Shan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Junjie Shen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanzhou Wang
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ping Yan
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Limei Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wenxu Zhao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanmin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wei Zhu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li Su
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Feifei Cheng
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hong Yao
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Huicheng Xu
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhiqing Liang
- Department of Obstetrics & Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
52
|
Nouri M, Caradec J, Lubik AA, Li N, Hollier BG, Takhar M, Altimirano-Dimas M, Chen M, Roshan-Moniri M, Butler M, Lehman M, Bishop J, Truong S, Huang SC, Cochrane D, Cox M, Collins C, Gleave M, Erho N, Alshalafa M, Davicioni E, Nelson C, Gregory-Evans S, Karnes RJ, Jenkins RB, Klein EA, Buttyan R. Therapy-induced developmental reprogramming of prostate cancer cells and acquired therapy resistance. Oncotarget 2017; 8:18949-18967. [PMID: 28145883 PMCID: PMC5386661 DOI: 10.18632/oncotarget.14850] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/16/2017] [Indexed: 01/01/2023] Open
Abstract
Treatment-induced neuroendocrine transdifferentiation (NEtD) complicates therapies for metastatic prostate cancer (PCa). Based on evidence that PCa cells can transdifferentiate to other neuroectodermally-derived cell lineages in vitro, we proposed that NEtD requires first an intermediary reprogramming to metastable cancer stem-like cells (CSCs) of a neural class and we demonstrate that several different AR+/PSA+ PCa cell lines were efficiently reprogrammed to, maintained and propagated as CSCs by growth in androgen-free neural/neural crest (N/NC) stem medium. Such reprogrammed cells lost features of prostate differentiation; gained features of N/NC stem cells and tumor-initiating potential; were resistant to androgen signaling inhibition; and acquired an invasive phenotype in vitro and in vivo. When placed back into serum-containing mediums, reprogrammed cells could be re-differentiated to N-/NC-derived cell lineages or return back to an AR+ prostate-like state. Once returned, the AR+ cells were resistant to androgen signaling inhibition. Acute androgen deprivation or anti-androgen treatment in serum-containing medium led to the transient appearance of a sub-population of cells with similar characteristics. Finally, a 132 gene signature derived from reprogrammed PCa cell lines distinguished tumors from PCa patients with adverse outcomes. This model may explain neural manifestations of PCa associated with lethal disease. The metastable nature of the reprogrammed stem-like PCa cells suggests that cycles of PCa cell reprogramming followed by re-differentiation may support disease progression and therapeutic resistance. The ability of a gene signature from reprogrammed PCa cells to identify tumors from patients with metastasis or PCa-specific mortality implies that developmental reprogramming is linked to aggressive tumor behaviors.
Collapse
Affiliation(s)
- Mannan Nouri
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Josselin Caradec
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Amy Anne Lubik
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Na Li
- Vancouver Prostate Centre, Vancouver, Canada
| | - Brett G Hollier
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | | | - Mengqian Chen
- Drug Discovery & Biomedical Sciences, South Carolina College of Pharmacy, Columbia, South Carolina, USA
| | | | | | - Melanie Lehman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | | | | | - Dawn Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Michael Cox
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Nicholas Erho
- GenomeDX Biosciences, Vancouver, Canada.,GenomeDX Biosciences, San Diego, California, USA
| | | | - Elai Davicioni
- GenomeDX Biosciences, Vancouver, Canada.,GenomeDX Biosciences, San Diego, California, USA
| | - Colleen Nelson
- Vancouver Prostate Centre, Vancouver, Canada.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Sheryl Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, Canada
| | | | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Ralph Buttyan
- Vancouver Prostate Centre, Vancouver, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
53
|
Liu Q, Tong D, Liu G, Xu J, Do K, Geary K, Zhang D, Zhang J, Zhang Y, Li Y, Bi G, Lan W, Jiang J. Metformin reverses prostate cancer resistance to enzalutamide by targeting TGF-β1/STAT3 axis-regulated EMT. Cell Death Dis 2017; 8:e3007. [PMID: 28837141 PMCID: PMC5596596 DOI: 10.1038/cddis.2017.417] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023]
Abstract
Although the newly developed second-generation anti-androgen drug enzalutamide can repress prostate cancer progression significantly, it only extends the survival of prostate cancer patients by 4–6 months mainly due to the occurrence of enzalutamide resistance. Most of the previous studies on AR antagonist resistance have been focused on AR signaling. Therefore, the non-AR pathways on enzalutamide resistance remain largely unknown. By using C4-2, CWR22Rv1 and LNCaP cell lines, as well as mice bearing CWR22Rv1 xenografts treated with either enzalutamide or metformin alone or in combination, we demonstrated that metformin is capable of reversing enzalutamide resistance and restores sensitivity of CWR22Rv1 xenografts to enzalutamide. We showed that metformin alleviated resistance to enzalutamide by inhibiting EMT. Furthermore, based on the effect of metformin on the activation of STAT3 and expression of TGF-β1, we propose that metformin exerts its effects by targeting the TGF-β1/STAT3 axis. These findings suggest that combination of metformin with enzalutamide could be a more efficacious therapeutic strategy for the treatment of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jing Xu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Khang Do
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Kyla Geary
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Jun Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yao Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Yaoming Li
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gang Bi
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
54
|
Chen J, Li L, Yang Z, Luo J, Yeh S, Chang C. Androgen-deprivation therapy with enzalutamide enhances prostate cancer metastasis via decreasing the EPHB6 suppressor expression. Cancer Lett 2017; 408:155-163. [PMID: 28826721 DOI: 10.1016/j.canlet.2017.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
Abstract
Early studies suggested that using ADT with the recently developed anti-androgen Enzalutamide (Enz, also named as MDV3100 could extent castration resistant prostate cancer (CRPC) patients' survival an extra 4.8 months. Yet the therapy in most patients might eventually fail due to development of Enz-resistance. Here we found Enz might also increase some unwanted side-effects via increasing the CRPC cell invasion that might involve altering the Enz-mediated androgen receptor (AR)/EPHB6 suppressor/JNK signaling. Results from multiple clinical surveys also indicated that EPHP6 might function as a suppressor of PCa metastasis. Mechanism dissection revealed that Enz-mediated AR might function via binding to the androgen-response-element (ARE) on the EPHB6 promoter to decrease EPHB6 suppressor expression, which might then activate the phosphorylation of JNK signals to increase the CRPC cell invasion. Targeting this newly identified AR/EPHB6/JNK signaling with JNK inhibitor (SP600125) may then block/reverse the Enz-increased CRPC cell invasion. Collectively, our results suggest that Enz may increase CRPC cell invasion via altering the AR/EPHB6/JNK/MMP9 signaling and targeting this newly identified signaling may help us to increase the Enz efficacy to better suppress the CRPC at the later metastatic stage.
Collapse
Affiliation(s)
- Jiaqi Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lei Li
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Zhao Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jie Luo
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Sex Hormone Research Center, China Medical University/Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
55
|
Targeting androgen receptor versus targeting androgens to suppress castration resistant prostate cancer. Cancer Lett 2017; 397:133-143. [DOI: 10.1016/j.canlet.2017.03.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/11/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022]
|
56
|
Rycaj K, Li H, Zhou J, Chen X, Tang DG. Cellular determinants and microenvironmental regulation of prostate cancer metastasis. Semin Cancer Biol 2017; 44:83-97. [PMID: 28408152 PMCID: PMC5491097 DOI: 10.1016/j.semcancer.2017.03.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022]
Abstract
Metastasis causes more than 90% of cancer-related deaths and most prostate cancer (PCa) patients also die from metastasis. The 'metastatic cascade' is a complex biological process that encompasses tumor cell dissociation (from the primary tumor), local invasion, intravasation, transport in circulation, extravasation, colonization, and overt growth in end organs. It has become clear that successful metastasis not only involves many tumor cell-intrinsic properties but also depends on productive interactions between cancer cells and the tumor microenvironment. In this Review, we begin with a general summary on cancer metastasis and a specific discussion on PCa metastasis. We then discuss recent advances in our knowledge of the cellular determinants of PCa metastasis and the importance of tumor microenvironment, especially an immunosuppressive tumor microenvironment, in shaping metastatic propensities. We conclude with a presentation of current and future therapeutic options for patients with PCa metastasis, emphasizing the development of novel, mechanism-based combinatorial strategies for treating metastatic and castration-resistant PCa.
Collapse
Affiliation(s)
- Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Hangwen Li
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jianjun Zhou
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xin Chen
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
57
|
Lin SJ, Chou FJ, Li L, Lin CY, Yeh S, Chang C. Natural killer cells suppress enzalutamide resistance and cell invasion in the castration resistant prostate cancer via targeting the androgen receptor splicing variant 7 (ARv7). Cancer Lett 2017; 398:62-69. [PMID: 28373004 DOI: 10.1016/j.canlet.2017.03.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 10/19/2022]
Abstract
Despite the success of androgen-deprivation therapy (ADT) with the newly developed anti-androgen enzalutamide (Enz, also known as MDV3100) to suppress castration resistant prostate cancer (CRPC) in extending patient survival by an extra 4.8 months, eventually patients die with the development of Enz resistance that may involve the induction of the androgen receptor (AR) splicing variant ARv7. Here we identify an unrecognized role of Natural Killer (NK) cells in the prostate tumor microenvironment that can be better recruited to the CRPC cells to suppress ARv7 expression resulting in suppressing the Enz resistant CRPC cell growth and invasion. Mechanism dissection revealed that CRPC cells, compared to normal prostate epithelial cells, could recruit more NK cells that might then lead to alterations of the microRNA-34 and microRNA-449 to suppress both ARv7 expression and ARv7-induced EZH2 expression to suppress CRPC cell invasion. Together, these results identify a new potential therapy using recruited NK cells to better suppress the Enz resistance and cell invasion in CRPC at the later enzalutamide resistant stage.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Fu-Ju Chou
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lei Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA; Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Chang-Yi Lin
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA; Sex Hormone Research Center, China Medical University/Hospital, Taichung 404, Taiwan.
| |
Collapse
|
58
|
Camora LF, Silva APG, Santos SAA, Justulin LA, Perobelli JE, Barbisan LF, Scarano WR. Impact of maternal and postnatal zinc dietary status on the prostate of pubescent and adult rats. Cell Biol Int 2017; 41:1203-1213. [PMID: 28244627 DOI: 10.1002/cbin.10756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/24/2017] [Indexed: 02/06/2023]
Abstract
Zinc is important for cell physiology and alteration of its levels during development can modulate a series of biological events. The aim of this study was to investigate whether dietary zinc deficiency or supplementation during morphogenesis and early postnatal development could interfere in prostate maturation. Pregnant rats were exposed to a standard diet (NZ:35 mg Zn/kg chow), low-zinc diet (LZ:3 mg of Zn/kg chow) and zinc-supplemented diet (HZ:180 mg/Kg chow) from gestational day 10 (GD10) through postnatal day 21 (PND21). After weaning, male offspring were divided into three groups that were submitted to the same food conditions as their mothers until PND53. The animals were euthanized at PND53 and PND115. The ventral prostate was removed, weighed and its fragments were subjected to histological, western blot and zymography analysis. PND53: body and prostate weight were lower in LZ compared to NZ; the epithelial compartment was reduced while the stromal compartment was increased in LZ compared to NZ; there was an increase in the amount of collagen and reduction in AR and SIRT1 expression in LZ compared to NZ. PND115: body weight was lower in LZ compared to NZ and prostate weight was similar among the groups; peripheral physiological hyperplasia was observed, as well as an increased epithelial proliferation index and reduced PAR4 expression in LZ and HZ compared to NZ. Zinc deficiency during prostate morphogenesis and differentiation is potentially harmful to its morphology, however, by restoring the standard dietary environment, the gland responds to the new microenvironment independent of the previous dietary condition.
Collapse
Affiliation(s)
- Lucas F Camora
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Ana Priscila G Silva
- Department of Sciences of the Sea, Federal University of São Paulo, UNIFESP, Santos, São Paulo, Brazil
| | - Sérgio A A Santos
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Luis A Justulin
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Juliana E Perobelli
- Department of Sciences of the Sea, Federal University of São Paulo, UNIFESP, Santos, São Paulo, Brazil
| | - Luis Fernando Barbisan
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Wellerson R Scarano
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| |
Collapse
|
59
|
Wang Y, Ou Z, Sun Y, Yeh S, Wang X, Long J, Chang C. Androgen receptor promotes melanoma metastasis via altering the miRNA-539-3p/USP13/MITF/AXL signals. Oncogene 2016; 36:1644-1654. [PMID: 27869170 DOI: 10.1038/onc.2016.330] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 07/10/2015] [Accepted: 07/28/2016] [Indexed: 12/21/2022]
Abstract
Early studies demonstrated that male melanoma patients have worse survival than female patients, yet the detailed mechanisms for this gender difference remain unclear. We analyzed around 100 cases of human melanoma and found that androgen receptor (AR) positive melanoma patients have worse survival outcomes compared with AR-negative melanoma patients. Here we report that AR can have positive roles to increase melanoma cell invasion in multiple cell lines in vitro and a mouse model in vivo. Mechanism dissection suggest that AR increases melanoma cell invasion via modulating the MITF-AXL signals via altering the miRNA-539-3p/USP13 signaling to increase MITF protein degradation through a reduction of de-ubiquitination. Restoring MITF can reverse AR-enhanced melanoma cell invasion. Together, our results demonstrate that AR can promote melanoma metastasis via altering the miRNA-539-3p/USP13/MITF/AXL signal and targeting this newly identified signal with AR degradation enhancer ASC-J9 may help us to better suppress the melanoma metastasis.
Collapse
Affiliation(s)
- Y Wang
- Departments of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.,George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester NY, USA
| | - Z Ou
- Departments of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.,George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester NY, USA
| | - Y Sun
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester NY, USA
| | - S Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester NY, USA
| | - X Wang
- Departments of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - J Long
- Departments of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - C Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester NY, USA.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
60
|
Xu J, Lin H, Li G, Sun Y, Shi L, Ma WL, Chen J, Cai X, Chang C. Sorafenib with ASC-J9 ® synergistically suppresses the HCC progression via altering the pSTAT3-CCL2/Bcl2 signals. Int J Cancer 2016; 140:705-717. [PMID: 27668844 PMCID: PMC5215679 DOI: 10.1002/ijc.30446] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 12/21/2022]
Abstract
Sorafenib is currently used as a standard treatment to suppress the progression of hepatocellular carcinoma (HCC), especially in advanced stages. However, patients who receive Sorafenib treatment eventually develop resistance without clear mechanisms. There is a great need for better efficacy of Sorafenib treatment in combination with other therapies. Here, we demonstrated that the treatment combining Sorafenib with ASC‐J9® could synergistically suppress HCC progression via altering cell‐cycle regulation, apoptosis and invasion. Mechanism dissection suggests that while Sorafenib impacts little or even slightly increases the activated/phosphorylated STAT3 (p‐STAT3), a key stimulator to promote the HCC progression, adding ASC‐J9® significantly suppresses the p‐STAT3 expression and its downstream genes including CCL2 and Bcl2. Interrupting these signals via constitutively active STAT3 partially reverses the synergistic suppression of Sorafenib‐ASC‐J9® combination on HCC progression. In vivo studies further confirmed the synergistic effect of Sorafenib‐ASC‐J9® combination. Together, these results suggest the newly developed Sorafenib‐ASC‐J9® combination is a novel therapy to better suppress HCC progression. What's new? Sorafenib is currently a standard treatment to suppress the progression of hepatocellular carcinoma (HCC). STAT3 activation may however play a role in the development of Sorafenib resistance. Following earlier studies suggesting that ASC‐J9® may alter activated p‐STAT3 signals to suppress prostate cancer metastasis, here the authors found that combining Sorafenib with ASC‐J9® may synergistically suppress HCC progression. Sorafenib had little impact on p‐STAT3, whereas ASC‐J9® significantly suppressed p‐STAT3 expression and its downstream genes, including CCL2 and Bcl2. Clinical studies using human HCC samples also demonstrated that higher expression of p‐STAT3 might be linked to the lower response to Sorafenib treatment.
Collapse
Affiliation(s)
- Junjie Xu
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Hui Lin
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Gonghui Li
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Liang Shi
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Wen-Lung Ma
- Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| | - Jiang Chen
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Xiujun Cai
- Chawnshang Chang Liver Cancer Center, Departments of General Surgery and Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
61
|
Di Zazzo E, Galasso G, Giovannelli P, Di Donato M, Di Santi A, Cernera G, Rossi V, Abbondanza C, Moncharmont B, Sinisi AA, Castoria G, Migliaccio A. Prostate cancer stem cells: the role of androgen and estrogen receptors. Oncotarget 2016; 7:193-208. [PMID: 26506594 PMCID: PMC4807992 DOI: 10.18632/oncotarget.6220] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men, and androgen deprivation therapy still represents the primary treatment for prostate cancer patients. This approach, however, frequently fails and patients develop castration-resistant prostate cancer, which is almost untreatable. Cancer cells are characterized by a hierarchical organization, and stem/progenitor cells are endowed with tumor-initiating activity. Accumulating evidence indicates that prostate cancer stem cells lack the androgen receptor and are, indeed, resistant to androgen deprivation therapy. In contrast, these cells express classical (α and/or β) and novel (GPR30) estrogen receptors, which may represent new putative targets in prostate cancer treatment. In the present review, we discuss the still-debated mechanisms, both genomic and non-genomic, by which androgen and estradiol receptors (classical and novel) mediate the hormonal control of prostate cell stemness, transformation, and the continued growth of prostate cancer. Recent preclinical and clinical findings obtained using new androgen receptor antagonists, anti-estrogens, or compounds such as enhancers of androgen receptor degradation and peptides inhibiting non-genomic androgen functions are also presented. These new drugs will likely lead to significant advances in prostate cancer therapy.
Collapse
Affiliation(s)
- Erika Di Zazzo
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Giovanni Galasso
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Pia Giovannelli
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Marzia Di Donato
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Annalisa Di Santi
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Gustavo Cernera
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Valentina Rossi
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Ciro Abbondanza
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | | | - Antonio Agostino Sinisi
- Endocrinology Section, Department of Cardio-Thoracic and Respiratory Diseases, II University of Naples, Naples, Italy
| | - Gabriella Castoria
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics and General Pathology, II University of Naples, Naples, Italy
| |
Collapse
|
62
|
Schmidt KT, Figg WD. The potential role of curcumin in prostate cancer: the importance of optimizing pharmacokinetics in clinical studies. Transl Cancer Res 2016; 5:S1107-S1110. [PMID: 30613476 DOI: 10.21037/tcr.2016.11.04] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Keith T Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
63
|
Nohara S, Kato K, Fujiwara D, Sakuragi N, Yanagihara K, Iwanuma Y, Kajiyama Y. Aminopeptidase N (APN/CD13) as a target molecule for scirrhous gastric cancer. Clin Res Hepatol Gastroenterol 2016; 40:494-503. [PMID: 26774363 PMCID: PMC7185882 DOI: 10.1016/j.clinre.2015.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 11/04/2015] [Accepted: 11/15/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Scirrhous gastric cancer is associated with peritoneal dissemination and advanced lymph node metastasis from an early stage, and the prognosis is still poor. In this study, we aimed to analyze candidate molecules for targeted therapy of scirrhous gastric cancer. We searched for molecules/metabolic activity that might be predominantly expressed in a subpopulation of scirrhous gastric cancer cells and might function as cancer stem cell markers. RESULTS For this purpose, we investigated the expression of various cell surface markers and of aldehyde dehydrogenase (ALDH) activity. These analyses showed that the scirrhous gastric cancer cell lines HSC-58 and HSC-44PE heterogeneously expressed CD13, while CD44, CDCP1, EpCAM and ABCG2 were expressed uniformly. Moreover, 10% of the total HSC-58 cell population expressed ALDH enzyme activity. A subpopulation of cells strongly positive for ALDH also expressed high levels of CD13, both of which are known as cancer stem cell markers. HSC-58 cells expressing high levels of CD13 showed lower sensitivity to a cancer drug cisplatin than cells with low levels of CD13. In contrast, CD13(-high) subpopulation of HSC-58 was more sensitive to an aminopeptidase N inhibitor bestatin. In terms of antibody-drug therapy, anti-CD13-immunotoxin was highly cytotoxic towards HSC-58 cells and was more cytotoxic than anti-EpCAM-immunotoxin. CONCLUSION These data suggest that CD13 is a suitable cell surface candidate for targeted antibody-drug therapy of scirrhous gastric cancer.
Collapse
Affiliation(s)
- Shigeo Nohara
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kazunori Kato
- Department of Biomedical Engineering, Toyo University, 2100 Kujirai, Kawagoe, Saitama 350-8585, Japan,Atopy Research Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan,Corresponding author. Department of Biomedical Engineering, Toyo University, 2100 Kujirai, Kawagoe, Saitama 350-8585, Japan.
| | - Daisuke Fujiwara
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Naoya Sakuragi
- Department of Biomedical Engineering, Toyo University, 2100 Kujirai, Kawagoe, Saitama 350-8585, Japan
| | - Kazuyoshi Yanagihara
- Division of Translational Research, Exploratory Oncology and Clinical Trial Center, National Cancer Center Hospital East, 6-5-1 Kashiwano-ha, Kashiwa, Chiba 277-8577, Japan
| | - Yoshimi Iwanuma
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshiaki Kajiyama
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
64
|
Infiltrating bone marrow mesenchymal stem cells (BM-MSCs) increase prostate cancer cell invasion via altering the CCL5/HIF2α/androgen receptor signals. Oncotarget 2016; 6:27555-65. [PMID: 26342197 PMCID: PMC4695008 DOI: 10.18632/oncotarget.4515] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/17/2015] [Indexed: 11/25/2022] Open
Abstract
Several infiltrating cells in the tumor microenvironment could influence the cancer progression via secreting various cytokines. Here, we found the CCL5 secreted from BM-MSCs suppressed androgen receptor (AR) signals via enhancing the expression of hypoxia inducible factor 2α (HIF2α) in prostate cancer (PCa) cells. Mechanism dissection revealed that the increased HIF2α might alter the AR-HSP90 interaction to suppress the AR transactivation, and inhibition of HIF2α reversed the BM-MSCs-increased PCa stem cell population and PCa cells invasion. Importantly, CCL5 could suppress the prolyl hydroxylases (PHDs) expression, which might then lead to suppress VHL-mediated HIF2α ubiquitination. Together, these results demonstrated that the CCL5 signals from infiltrating BM-MSC cells to HIF2α signals within PCa cells might play a key role to increase PCa stem cell population and PCa metastasis via altering the AR signals. Targeting this newly identified CCL5/HIF2α/AR axis signal axis may allow us to develop a novel way to suppress PCa metastasis.
Collapse
|
65
|
Wen S, Tian J, Niu Y, Li L, Yeh S, Chang C. ASC-J9®, and not Casodex or Enzalutamide, suppresses prostate cancer stem/progenitor cell invasion via altering the EZH2-STAT3 signals. Cancer Lett 2016; 376:377-86. [DOI: 10.1016/j.canlet.2016.01.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/03/2015] [Accepted: 01/11/2016] [Indexed: 11/27/2022]
|
66
|
ASC-J9(®) suppresses castration resistant prostate cancer progression via degrading the enzalutamide-induced androgen receptor mutant AR-F876L. Cancer Lett 2016; 379:154-60. [PMID: 27233475 DOI: 10.1016/j.canlet.2016.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/27/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Androgen deprivation therapy (ADT) with the newly developed powerful anti-androgen enzalutamide (Enz, also known as MDV3100) has promising therapeutic effects to suppress castration resistant prostate cancer (CRPC) and extending patients' lives an extra 4.8 months. However, most Enz therapy eventually fails with the development of Enz resistance. The detailed mechanisms how CRPC develops Enz resistance remain unclear and may involve multiple mechanisms. Among them, the induction of the androgen receptor (AR) mutant AR-F876L in some CRPC patients may represent one driving force that confers Enz resistance. Here, we demonstrate that the AR degradation enhancer, ASC-J9(®), not only degrades wild-type AR, but also has the ability to target AR-F876L. The consequence of suppressing AR-F876L may then abrogate AR-F876L mediated CRPC cell proliferation and metastasis. Thus, developing ASC-J9(®) as a new therapeutic approach may represent a novel therapy to better suppress CRPC that has already developed Enz resistance.
Collapse
|
67
|
Li L, Dang Q, Xie H, Yang Z, He D, Liang L, Song W, Yeh S, Chang C. Infiltrating mast cells enhance prostate cancer invasion via altering LncRNA-HOTAIR/PRC2-androgen receptor (AR)-MMP9 signals and increased stem/progenitor cell population. Oncotarget 2016; 6:14179-90. [PMID: 25895025 PMCID: PMC4546459 DOI: 10.18632/oncotarget.3651] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/28/2015] [Indexed: 12/14/2022] Open
Abstract
Early studies indicated that selective inflammatory immune cells in the prostate tumor microenvironment might be able to influence prostate cancer (PCa) progression. Here we found treating PCa cells with androgen deprivation therapy (ADT) results in the recruitment of more mast cells, which might then increase PCa cell invasion via down-regulation of AR signals in 4 different PCa cell lines. Mechanism dissection revealed infiltrating mast cells could decrease AR transcription via modulation of the PRC2 complex with LncRNA-HOTAIR at the AR 5' promoter region in PCa cells. The consequences of suppressing AR may then increase PCa cell invasion via increased MMP9 expression and/or increased stem/progenitor cell population. The in vivo mouse model with orthotopically xenografted PCa CWR22Rv1 cells with/without mast cells also confirmed that infiltrating mast cells could increase PCa cell invasion via suppression of AR signals. Together, our results provide a new mechanism for the ADT-enhanced PCa metastasis via altering the infiltrating mast cells to modulate PCa AR-MMP9 signals and/or AR-stem/progenitor cell population. Targeting these newly identified inflammatory mast cells-AR signals may help us to better suppress PCa metastasis at the castration resistant stage.
Collapse
Affiliation(s)
- Lei Li
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Qiang Dang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Hongjun Xie
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Zhao Yang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dalin He
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Liang
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Wenbing Song
- Department of Urology, Sex Hormone Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Shuyuan Yeh
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
68
|
Abstract
Tumour metastasis, the movement of tumour cells from a primary site to progressively colonize distant organs, is a major contributor to the deaths of cancer patients. Therapeutic goals are the prevention of an initial metastasis in high-risk patients, shrinkage of established lesions and prevention of additional metastases in patients with limited disease. Instead of being autonomous, tumour cells engage in bidirectional interactions with metastatic microenvironments to alter antitumour immunity, the extracellular milieu, genomic stability, survival signalling, chemotherapeutic resistance and proliferative cycles. Can targeting of these interactions significantly improve patient outcomes? In this Review preclinical research, combination therapies and clinical trial designs are re-examined.
Collapse
Affiliation(s)
- Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
69
|
Nastiuk KL, Krolewski JJ. Opportunities and challenges in combination gene cancer therapy. Adv Drug Deliv Rev 2016; 98:35-40. [PMID: 26724249 PMCID: PMC4957561 DOI: 10.1016/j.addr.2015.12.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/28/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023]
Abstract
Treatment for solid tumor malignancies, which constitute the majority of human cancers, is still dominated by surgery and radiotherapies. This is especially true for many localized solid tumors, which are often curable with these treatments. However, metastatic cancers are beyond the reach of these therapies, and many localized cancers that are initially treated with surgery and radiation will recur and metastasize. Thus, for over 60years there has been a concerted effort to develop effective drug treatments for metastatic cancers. Combination therapies are an increasingly important part of the anti-cancer drug armamentarium. In the case of cytotoxic chemotherapy, multi-drug regimens rapidly became the norm, as the earliest single agents were relatively ineffective. In contrast to chemotherapy, where combination therapies were required in order to achieve treatment efficacy, for both hormonal and targeted therapies the impetus to move toward the use of combination therapies is to prevent or reverse the development of treatment resistance. In addition, emerging evidence suggests that combination therapy may also improve cancer treatment by neutralizing an emerging treatment side effect termed therapy-induced metastasis, which accompanies some effective single agent therapies. Finally, although gene therapy is still far from use in the clinic, we propose that combination therapies may enhance its effectiveness.
Collapse
Affiliation(s)
- Kent L Nastiuk
- Department of Cancer Genetics, Elm & Carlton Streets, Buffalo, NY 14263, United States
| | - John J Krolewski
- Roswell Park Cancer Institute, Center for Personalized Medicine, United States; Department of Cancer Genetics, Elm & Carlton Streets, Buffalo, NY 14263, United States.
| |
Collapse
|
70
|
Wen S, Niu Y, Lee SO, Yeh S, Shang Z, Gao H, Li Y, Chou F, Chang C. Targeting fatty acid synthase with ASC-J9 suppresses proliferation and invasion of prostate cancer cells. Mol Carcinog 2016; 55:2278-2290. [PMID: 26894509 DOI: 10.1002/mc.22468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 01/09/2016] [Accepted: 01/15/2016] [Indexed: 01/22/2023]
Abstract
Fatty acid synthase (FASN) is the key enzyme for the control of fatty acid synthesis that contributes significantly to the prostate cancer (PCa) progression. It was reported that androgens were able to induce FASN expression in PCa, and addition of the anti-androgen Casodex might suppress the androgen-induced FASN expression. However, here we found androgen-deprivation-therapy (ADT) with anti-androgens Bicalutamide (Casodex) or Enzalutamide (MDV3100) had little effect to suppress FASN expression and FASN-mediated cell growth and invasion during the castration resistant stage when the androgen concentration is 1 nM DHT (dihydrotestosterone). In contrast, the newly developed androgen receptor (AR) degradation enhancer ASC-J9® suppressed FASN expression and FASN-mediated cell growth and invasion in various PCa cell lines at 1 nM DHT. Mechanism dissection found ASC-J9® could suppress significantly the FASN expression and FASN-mediated PCa progression via the AR-dependent pathway involving AR→SREBP-1→FASN signaling in AR-positive C4-2 and LNCaP cells and via the AR-independent pathway involving the modulation of PI3K/AKT→SREBP-1→FASN signaling in AR-negative PC-3 and DU145 cells. Together, these results suggest that FASN is one of the important mechanism why the current ADT eventually fails. ASC-J9® might represent a new potential therapeutic approach to suppress FASN-mediated PCa progression via both AR-dependent and AR-independent pathways during the castration resistant stage of PCa. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simeng Wen
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Soo Ok Lee
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Shuyuan Yeh
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Zhiqun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Hengheng Gao
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Yanjun Li
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Fuju Chou
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
71
|
Huang CK, Luo J, Lee SO, Chang C. Concise review: androgen receptor differential roles in stem/progenitor cells including prostate, embryonic, stromal, and hematopoietic lineages. Stem Cells 2015; 32:2299-308. [PMID: 24740898 DOI: 10.1002/stem.1722] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 01/07/2023]
Abstract
Stem/progenitor (S/P) cells are special types of cells that have the ability to generate tissues throughout their entire lifetime and play key roles in the developmental process. Androgen and the androgen receptor (AR) signals are the critical determinants in male gender development, suggesting that androgen and AR signals might modulate the behavior of S/P cells. In this review, we summarize the AR effects on the behavior of S/P cells, including self-renewal, proliferation, apoptosis, and differentiation in normal S/P cells, as well as proliferation, invasion, and self-renewal in prostate cancer S/P cells. AR plays a protective role in the oxidative stress-induced apoptosis in embryonic stem cells. AR inhibits the self-renewal of embryonic stem cells, bone marrow stromal cells, and prostate S/P cells, but promotes their differentiation except for adipogenesis. However, AR promotes the proliferation of hematopoietic S/P cells and stimulates hematopoietic lineage differentiation. In prostate cancer S/P cells, AR suppresses their self-renewal, metastasis, and invasion. Together, AR differentially influences the characteristics of normal S/P cells and prostate cancer S/P cells, and targeting AR might improve S/P cell transplantation therapy, especially in embryonic stem cells and bone marrow stromal cells.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Departments of Pathology, Urology, Radiation Oncology, the George Whipple Lab for Cancer Research, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | |
Collapse
|
72
|
Prostate Cancer Stem-like Cells Contribute to the Development of Castration-Resistant Prostate Cancer. Cancers (Basel) 2015; 7:2290-308. [PMID: 26593949 PMCID: PMC4695890 DOI: 10.3390/cancers7040890] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/30/2015] [Accepted: 11/04/2015] [Indexed: 12/16/2022] Open
Abstract
Androgen deprivation therapy (ADT) has been the standard care for patients with advanced prostate cancer (PC) since the 1940s. Although ADT shows clear benefits for many patients, castration-resistant prostate cancer (CRPC) inevitably occurs. In fact, with the two recent FDA-approved second-generation anti-androgens abiraterone and enzalutamide, resistance develops rapidly in patients with CRPC, despite their initial effectiveness. The lack of effective therapeutic solutions towards CRPC largely reflects our limited understanding of the underlying mechanisms responsible for CRPC development. While persistent androgen receptor (AR) signaling under castration levels of serum testosterone (<50 ng/mL) contributes to resistance to ADT, it is also clear that CRPC evolves via complex mechanisms. Nevertheless, the physiological impact of individual mechanisms and whether these mechanisms function in a cohesive manner in promoting CRPC are elusive. In spite of these uncertainties, emerging evidence supports a critical role of prostate cancer stem-like cells (PCSLCs) in stimulating CRPC evolution and resistance to abiraterone and enzalutamide. In this review, we will discuss the recent evidence supporting the involvement of PCSLC in CRPC acquisition as well as the pathways and factors contributing to PCSLC expansion in response to ADT.
Collapse
|
73
|
Sha K, Yeh S, Chang C, Nastiuk KL, Krolewski JJ. TNF signaling mediates an enzalutamide-induced metastatic phenotype of prostate cancer and microenvironment cell co-cultures. Oncotarget 2015; 6:25726-40. [PMID: 26327448 PMCID: PMC4694862 DOI: 10.18632/oncotarget.4535] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/17/2015] [Indexed: 12/25/2022] Open
Abstract
The dramatic responses tumors display to targeted therapies are limited by acquired or pre-existing mechanisms of therapy resistance. We recently discovered that androgen receptor blockade by the anti-androgen enzalutamide paradoxically enhanced metastasis and that these pro-metastatic effects were mediated by the chemoattractant CCL2. CCL2 is regulated by TNF, which is negatively regulated by androgen signaling. Thus, we asked if TNF mediates the pro-metastatic effects of enzalutamide. We found that androgen withdrawal or enzalutamide induced TNF mRNA and protein secretion in castration resistant prostate cancer (C4-2) cells, but not in macrophage-like (THP1) or myofibroblast-like (WPMY1) cells. Androgen deprivation therapy (ADT) induced autocrine CCL2 expression in C4-2 (as well as a murine CRPC cell line), while exogenous TNF induced CCL2 in THP1 and WPMY1. TNF was most potent in myofibroblast cultures, suggesting ADT induces CCL2 via paracrine interactions within the tumor microenvironment. A soluble TNF receptor (etanercept) blocked enzalutamide-induced CCL2 protein secretion and mRNA, implying dependence on secreted TNF. A small molecule inhibitor of CCR2 (the CCL2 receptor) significantly reduced TNF induced migration, while etanercept inhibited enzalutamide-induced migration and invasion of C4-2. Analysis of human prostate cancers suggests that a TNF-CCL2 paracrine loop is induced in response to ADT and might account for some forms of prostate cancer therapy resistance.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Benzamides
- Cell Line, Tumor
- Cell Movement/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Coculture Techniques
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Myofibroblasts/pathology
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Nitriles
- Paracrine Communication/drug effects
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, CCR2/antagonists & inhibitors
- Receptors, CCR2/metabolism
- Signal Transduction/drug effects
- Time Factors
- Tumor Microenvironment/drug effects
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Up-Regulation
Collapse
Affiliation(s)
- Kai Sha
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Shuyuan Yeh
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Urology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
| | - Chawnshang Chang
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Urology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Radiation Oncology, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
| | - Kent L. Nastiuk
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - John J. Krolewski
- Department of Pathology and Laboratory Medicine, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester, School of Medicine and Dentistry; Rochester, NY 14642, USA
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263
| |
Collapse
|
74
|
Liu C, Lou W, Armstrong C, Zhu Y, Evans CP, Gao AC. Niclosamide suppresses cell migration and invasion in enzalutamide resistant prostate cancer cells via Stat3-AR axis inhibition. Prostate 2015; 75:1341-53. [PMID: 25970160 PMCID: PMC4536195 DOI: 10.1002/pros.23015] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/16/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE It is known that over expression of IL6 in prostate cancer cells confer enzalutamide resistance and that this may occur through constitutive Stat3 activation. Additionally, recent pre-clinical studies suggested enzalutamide might have the potential adverse effect of inducing metastasis of prostate cancer cells via Stat3 activation. This study is aimed to target Stat3 activation and improve enzalutamide therapy. EXPERIMENTAL DESIGN Sensitivity of prostate cancer cells to enzalutamide was tested using cell growth assays and clonogenic assays. Wound healing and invasion assays were performed to determine cell migration and invasion in vitro. Quantitative reverse transcription-PCR, ELISA and Western blotting were performed to detect expression levels of PSA, c-Myc, survivin, Stat3, and AR. ChIP assay was performed to examine recruitment of AR to the PSA promoter. RESULTS In the present study, we found niclosamide, a previously identified novel inhibitor of androgen receptor variant (AR-V7), inhibited Stat3 phosphorylation, and expression of downstream target genes. Niclosamide synergistically reversed enzalutamide resistance in prostate cancer cells and combination treatment of niclosamide with enzalutamide significantly induced cell apoptosis and inhibited cell growth, colony formation, cell migration and invasion. Knock down of Stat3 abrogated enzalutamide resistance resulting in reduced recruitment of AR to the PSA promoter in prostate cancer cells expressing IL6. Moreover, niclosamide reversed enzalutamide resistance by down-regulating Stat3 target gene expression Stat3and abrogating recruitment of AR to PSA promoter resulting in PSA inhibition. CONCLUSIONS This study demonstrated the IL6-Stat3-AR axis in prostate cancer is one of the crucial mechanisms of enzalutamide resistance. Niclosamide has the potential to target the IL6-Stat3-AR pathway to overcome enzalutamide resistance and inhibit migration and invasion in advanced prostate cancer.
Collapse
Affiliation(s)
- Chengfei Liu
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - Wei Lou
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - Cameron Armstrong
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - Yezi Zhu
- Department of Urology, University of California at Davis, Sacramento, CA, USA
- Graduate Program in Pharmacology and Toxicology, University of California at Davis, Sacramento, CA, USA
| | - Christopher P Evans
- Department of Urology, University of California at Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California at Davis, Sacramento, CA, USA
| | - Allen C. Gao
- Department of Urology, University of California at Davis, Sacramento, CA, USA
- Graduate Program in Pharmacology and Toxicology, University of California at Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California at Davis, Sacramento, CA, USA
- To whom correspondence should be addressed: Department of Urology University of California Davis Medical Center 4645 2 Ave, Research III, Suite 1300 Sacramento, CA 95817
| |
Collapse
|
75
|
Katzenwadel A, Wolf P. Androgen deprivation of prostate cancer: Leading to a therapeutic dead end. Cancer Lett 2015; 367:12-7. [PMID: 26185001 DOI: 10.1016/j.canlet.2015.06.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/16/2015] [Accepted: 06/29/2015] [Indexed: 11/19/2022]
Abstract
Androgen deprivation therapy (ADT) is considered as the standard therapy for men with de novo or recurrent metastatic prostate cancer. ADT commonly leads to initial biochemical and clinical responses. However, several months after the beginning of treatment, tumors become castration-resistant and virtually all patients show disease progression. At this stage, tumors are no longer curable and cancer treatment options are only palliative. In this review, we describe molecular alterations in tumor cells during ADT, which lead to deregulation of different signaling pathways and castration-resistance, and how they might interfere with the clinical outcome of different second-line therapeutics. A recent breakthrough finding that early chemotherapy is associated with a significant survival benefit in metastatic hormone-sensitive disease highlights the fact that there is time for a fundamental paradigm shift in the treatment of advanced prostate cancer. Therapeutic intervention seems to be indicated before a castration-resistant stage is reached to improve therapeutic outcome and to reduce undesirable side effects.
Collapse
Affiliation(s)
- Arndt Katzenwadel
- Department of Urology, Medical Center, University of Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center, University of Freiburg, Engesser Strasse 4b, D-79108 Freiburg, Germany.
| |
Collapse
|
76
|
Dual inhibition of survivin and MAOA synergistically impairs growth of PTEN-negative prostate cancer. Br J Cancer 2015; 113:242-51. [PMID: 26103574 PMCID: PMC4506394 DOI: 10.1038/bjc.2015.228] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/12/2015] [Accepted: 05/28/2015] [Indexed: 01/20/2023] Open
Abstract
Background: Survivin and monoamine oxidase A (MAOA) levels are elevated in prostate cancer (PCa) compared to normal prostate glands. However, the relationship between survivin and MAOA in PCa is unclear. Methods: We examined MAOA expression in the prostate lobes of a conditional PTEN-deficient mouse model mirroring human PCa, with or without survivin knockout. We also silenced one gene at a time and examined the expression of the other. We further evaluated the combination of MAOA inhibitors and survivin suppressants on the growth, viability, migration and invasion of PCa cells. Results: Survivin and MAOA levels are both increased in clinical PCa tissues and significantly associated with patients' survival. Survivin depletion delayed MAOA increase during PCa progression, and silencing MAOA decreased survivin expression. The combination of MAOA inhibitors and the survivin suppressants (YM155 and SC144) showed significant synergy on the inhibition of PCa cell growth, migration and invasion with concomitant decrease in survivin and MMP-9 levels. Conclusions: There is a positive feedback loop between survivin and MAOA expression in PCa. Considering that survivin suppressants and MAOA inhibitors are currently available in clinical trials and clinical use, their synergistic effects in PCa support a rapid translation of this combination to clinical practice.
Collapse
|
77
|
Qiu X, Zhu J, Sun Y, Fan K, Yang DR, Li G, Yang G, Chang C. TR4 nuclear receptor increases prostate cancer invasion via decreasing the miR-373-3p expression to alter TGFβR2/p-Smad3 signals. Oncotarget 2015; 6:15397-409. [PMID: 25980442 PMCID: PMC4558159 DOI: 10.18632/oncotarget.3778] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/14/2015] [Indexed: 01/07/2023] Open
Abstract
Testicular nuclear receptor 4 (TR4), a member of the nuclear receptor superfamily, may play important roles to modulate the metabolic diseases and prostate tumorigenesis. Here we found TR4 could increase prostate cancer (PCa) cell invasion. Mechanism dissection revealed that TR4 might increase PCa cell invasion via decreasing the miR-373-3p expression that resulted in the activation of the TGFβR2/p-Smad3 signals. The in vivo mouse model using orthotopically xenografted CWR22Rv1 cell line transfected with luciferase-reporter confirmed in vitro cell line studies showing TR4 increased PCa metastasis via decreasing the miR-373-3p expression. Together, these data suggest that TR4 may increase PCa metastasis via a newly identified signal and targeting these TR4/miR-473-3p/TGFβR2/p-Smad3 signals using TR4 antagonist or TR4-siRNA or miR-373-3p may allow us to develop a new potential therapeutic approach to better suppress PCa metastasis.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation/genetics
- Cell Transformation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Mice, Nude
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- Neoplasm Invasiveness/pathology
- Neoplasm Transplantation
- Prostatic Neoplasms/pathology
- Protein Serine-Threonine Kinases/metabolism
- RNA Interference
- RNA, Small Interfering
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/metabolism
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction/genetics
- Smad3 Protein/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Xiaofu Qiu
- Department of Urology, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Jin Zhu
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Kun Fan
- Department of Urology, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Dong-Rong Yang
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gonghui Li
- Chawnshang Chang Liver Cancer Center, Department of Urology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Guosheng Yang
- Department of Urology, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
78
|
Zheng L, Chen J, Ma Z, Liu W, Yang F, Yang Z, Wang K, Wang X, He D, Li L. Capsaicin causes inactivation and degradation of the androgen receptor by inducing the restoration of miR-449a in prostate cancer. Oncol Rep 2015; 34:1027-34. [PMID: 26081756 DOI: 10.3892/or.2015.4055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/27/2015] [Indexed: 11/06/2022] Open
Abstract
Capsaicin, a novel antitumor agent extracted from chili peppers, has been proven to induce growth inhibition in various types of cancer including prostate cancer. However, the detailed mechanisms remain largely undiscovered. In the present study, we explored the regulation of the androgen receptor (AR) by capsaicin and further researched the mechanisms of their interaction in AR-positive prostate cancer cells. In the present study, cell viability was assessed by MTT assay. Cell cycle distribution was determined using flow cytometry. Expression levels of cyclin D1, miR-449a, AR and prostate-specific antigen (PSA) were assessed by quantitative real-time polymerase chain reaction or western blot analysis. To further confirm the relationship among miR-449a, AR and prostate cancer proliferation, miR-449a was overexpressed by a lentivirus in prostate cancer cells. We discovered that capsaicin prevented tumor proliferation and cell cycle progression through inactivation and degradation of AR. We also found that restoration of miR-449a induced by capsaicin treatment resulted in the inhibition of AR signaling. Finally, we demonstrated that increased expression of miR-449a sensitized prostate cancer to capsaicin treatment. Finally, our experimental results indicated that capsaicin negatively modulates the activity of AR at the mRNA and protein levels by restoring miR-449a profiling in prostate cancer. In addition, increased expression of miR-449a may facilitate the sensitivity of prostate cancer to capsaicin treatment. Thus, capsaicin may be developed as a novel anti-AR drug for the therapy of prostate cancer.
Collapse
Affiliation(s)
- Long Zheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Jiaqi Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Zhenkun Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Wei Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Fei Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Zhao Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
79
|
Bishop JL, Davies A, Ketola K, Zoubeidi A. Regulation of tumor cell plasticity by the androgen receptor in prostate cancer. Endocr Relat Cancer 2015; 22:R165-82. [PMID: 25934687 DOI: 10.1530/erc-15-0137] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2015] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) has become the most common form of cancer in men in the developed world, and it ranks second in cancer-related deaths. Men that succumb to PCa have a disease that is resistant to hormonal therapies that suppress androgen receptor (AR) signaling, which plays a central role in tumor development and progression. Although AR continues to be a clinically relevant therapeutic target in PCa, selection pressures imposed by androgen-deprivation therapies promote the emergence of heterogeneous cell populations within tumors that dictate the severity of disease. This cellular plasticity, which is induced by androgen deprivation, is the focus of this review. More specifically, we address the emergence of cancer stem-like cells, epithelial-mesenchymal or myeloid plasticity, and neuroendocrine transdifferentiation as well as evidence that demonstrates how each is regulated by the AR. Importantly, because all of these cell phenotypes are associated with aggressive PCa, we examine novel therapeutic approaches for targeting therapy-induced cellular plasticity as a way of preventing PCa progression.
Collapse
Affiliation(s)
- Jennifer L Bishop
- The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Alastair Davies
- The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Kirsi Ketola
- The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada The Vancouver Prostate Centre2660 Oak Street, Vancouver, British Columbia, Canada V6H-3Z6Department of Urologic SciencesUniversity of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
80
|
Li J, Xiang S, Zhang Q, Wu J, Tang Q, Zhou J, Yang L, Chen Z, Hann SS. Combination of curcumin and bicalutamide enhanced the growth inhibition of androgen-independent prostate cancer cells through SAPK/JNK and MEK/ERK1/2-mediated targeting NF-κB/p65 and MUC1-C. J Exp Clin Cancer Res 2015; 34:46. [PMID: 25971429 PMCID: PMC4446835 DOI: 10.1186/s13046-015-0168-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/08/2015] [Indexed: 11/17/2022] Open
Abstract
Background Prostate cancer is one of the most common malignancies in men. The mucin 1 (MUC1) heterodimeric oncoprotein is overexpressed in human prostate cancers with aggressive pathologic and clinical features, resulting in a poor outcome. However, the functional role for MUC1 C-terminal domain (MUC1-C) in androgen-independent prostate cancer occurrence and development has remained unclear. Methods Cell viability was measured by MTT assays. Western blot analysis was performed to measure the phosphorylation and protein expression of SAPK/JNK and ERK1/2, and MUC1-C, NF-κB subunit p65 and p50. Exogenous expression of MUC1-C, NF-κB subunit p65 was carried out by transient and electroporated transfection assays. Results We showed that curcumin inhibited the growth of androgen-independent prostate cancer cells and a synergy was observed in the presence of curcumin and bicalutamide, the androgen receptor antagonist. To further explore the potential mechanism underlining this, we found that curcumin increased the phosphorylation of ERK1/2 and SAPK/JNK, which was enhanced by bicalutamide. In addition, curcumin reduced the protein expression of MUC1-C and NF-κB subunit p65, which were abrogated in the presence of the inhibitors of MEK/ERK1/2 (PD98059) and SAPK/JNK (SP60015). A further reduction was observed in the combination of curcumin with bicalutamide. Moreover, while exogenous expression of MUC1-C had little effect on curcumin-reduced p65, the overexpression of p65 reversed the effect of curcumin on MUC1-C protein expression suggesting that p65 is upstream of MUC1-C. Intriguingly, we showed that exogenous expression of MUC1-C feedback diminished the effect of curcumin on phosphorylation of ERK1/2 and SAPK/JNK, and antagonized the effect of curcumin on cell growth. Conclusion Our results show that curcumin inhibits the growth of androgen-independent prostate cancer cells through ERK1/2- and SAPK/JNK-mediated inhibition of p65, followed by reducing expression of MUC1-C protein. More importantly, there are synergistic effects of curcumin and bicalutamide. The negative feedback regulatory loop of MUC1-C to ERK1/2 and SAPK/JNK further demonstrates the role of MUC1-C that contributes to the overall responses of curcumin. This study unveils the potential molecular mechanism by which combination of curcumin with bicalutamide enhances the growth inhibition of androgen-independent prostate cancer cells.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China. .,Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - SongTao Xiang
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - QiouHong Zhang
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - JingJing Wu
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Qing Tang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - JianFu Zhou
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - LiJun Yang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - ZhiQiang Chen
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China. .,Higher Education Mega Center, Panyu District, Guangdong Provincial Hospital of Chinese Medicine, No. 55, Neihuan West Road, Guangzhou, Guangdong Province, 510006, People's Republic of China.
| |
Collapse
|
81
|
Wang C, Sun H, Zou R, Zhou T, Wang S, Sun S, Tong C, Luo H, Li Y, Li Z, Wang E, Chen Y, Cao L, Li F, Zhao Y. MDC1 functionally identified as an androgen receptor co-activator participates in suppression of prostate cancer. Nucleic Acids Res 2015; 43:4893-908. [PMID: 25934801 PMCID: PMC4446443 DOI: 10.1093/nar/gkv394] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 04/14/2015] [Indexed: 02/06/2023] Open
Abstract
Mediator of DNA damage checkpoint protein 1 (MDC1) is essential for DNA damage response. However, the role of MDC1 in modulating gene transcription independently of DNA damage and the underlying mechanisms have not been fully defined. Androgen receptor (AR) is the central signaling pathway in prostate cancer (PCa) and its target genes are involved in both promotion and suppression of PCa. Here, we functionally identified MDC1 as a co-activator of AR. We demonstrate that MDC1 facilitates the association between AR and histone acetyltransferase GCN5, thereby increasing histone H3 acetylation level on cis-regulatory elements of AR target genes. MDC1 knockdown promotes PCa cells growth and migration. Moreover, depletion of MDC1 results in decreased expression of a subset of the endogenous androgen-induced target genes, including cell cycle negative regulator p21 and PCa metastasis inhibitor Vinculin, in AR positive PCa cell lines. Finally, the expression of MDC1 and p21 correlates negatively with aggressive phenotype of clinical PCa. These studies suggest that MDC1 as an epigenetic modifier regulates AR transcriptional activity and MDC1 may function as a tumor suppressor of PCa, and provide new insight into co-factor-AR-signaling pathway mechanism and a better understanding of the function of MDC1 on PCa.
Collapse
Affiliation(s)
- Chunyu Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongmiao Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Renlong Zou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Shengli Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Shiying Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Changci Tong
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hao Luo
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yanshu Li
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhenhua Li
- Department of Urology, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Enhua Wang
- Department of Pathology, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, China
| | - Yuhua Chen
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Liu Cao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Feng Li
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yue Zhao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
82
|
Abstract
Few pharmacotherapies are currently available to treat castration resistant prostate cancer (CRPC), with low impact on patient survival. Transforming growth factor-β (TGF-β) is a multi-functional peptide with opposite roles in prostate tumorigenesis as an inhibitor in normal growth and early stage disease and a promoter in advanced prostate cancer. Dysregulated TGF-β signaling leads to a cascade of events contributing to oncogenesis, including up-regulated proliferation, decreased apoptosis, epithelial-to-mesenchymal transition (EMT) and evasion of immune surveillance. TGF-β signaling pathway presents an appropriate venue for establishing a therapeutic targeting platform in CRPC. Exploitation of TGF-β effectors and their cross talk with the androgen axis pathway will provide new insights into mechanisms of resistance of the current antiandrogen therapeutic strategies and lead to generation of new effective treatment modalities for CRPC. Points of functional convergence of TGF-β with key oncogenic pathways, including mitogen-activated protein kinase (MAPK) and androgen receptor (AR), are discussed as navigated within the EMT landscape in the tumor microenvironment. In this context the emerging anti-TGF-β pharmacotherapies for prostate cancer treatment are considered. Targeting the functional cross-talk between the TGF-β signaling effectors with the androgen axis supports the development of novel therapeutic strategies for treating CRPC with high specificity and efficacy in a personalized-medicine approach.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Urology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Natasha Kyprianou
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Urology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
83
|
Dang Q, Li L, Xie H, He D, Chen J, Song W, Chang LS, Chang HC, Yeh S, Chang C. Anti-androgen enzalutamide enhances prostate cancer neuroendocrine (NE) differentiation via altering the infiltrated mast cells → androgen receptor (AR) → miRNA32 signals. Mol Oncol 2015; 9:1241-51. [PMID: 25817444 DOI: 10.1016/j.molonc.2015.02.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 10/23/2022] Open
Abstract
The recently developed anti-androgen enzalutamide also known as (MDV3100) has the advantage to prolong by 4.8 months the survival of castration resistant prostate cancer (CRPC) patients. However, the mechanisms behind the potential side effects involving the induction of the prostate cancer (PCa) neuroendocrine (NE) differentiation remain unclear. Here we found PCa cells could recruit more mast cells than normal prostate epithelial cells, and enzalutamide (or casodex) treatment could further increase such recruitment that resulted in promoting the PCa NE differentiation. Mechanism dissection found infiltrated mast cells could function through positive feedback to enhance PCa to recruit more mast cells via modulation of the androgen receptor (AR) → cytokines IL8 signals, and interruption by AR-siRNA or neutralizing anti-IL8 antibody could partially reverse the recruitment of mast cells. Importantly, targeting the PCa androgens/AR signals with AR-siRNA or enzalutamide (or casodex) also increased PCa NE differentiation via modulation of the miRNA32 expression, and adding miRNA32 inhibitor reversed the AR-siRNA- or enzalutamide-enhanced NE differentiation. Together, these results not only identified a new signal via infiltrated mast cells → PCa AR → miRNA32 to increase PCa NE differentiation, it also pointed out the potential unwanted side effects of enzalutamide (or casodex) to increase PCa NE differentiation. Targeting these newly identified signals, including AR, IL8, or miRNA32, may help us to better suppress PCa NE differentiation that is induced during ADT with anti-androgen enzalutamide (or casodex) treatment.
Collapse
Affiliation(s)
- Qiang Dang
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lei Li
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Hongjun Xie
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dalin He
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiaqi Chen
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenbing Song
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Luke S Chang
- Sex Hormone Research Center, Department of Urology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Hong-Chiang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA; Sex Hormone Research Center, China Medical University/Hospital, Taichung 404, Taiwan.
| |
Collapse
|
84
|
Wen SM, Quan CY, Jiang N, Shang ZQ, Niu YJ. What is the next generation therapeutic strategy for castration-resistant prostate cancer. Asian J Androl 2015; 17:223-4. [PMID: 25432503 PMCID: PMC4650465 DOI: 10.4103/1008-682x.143311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers in the world. Since androgen receptor (AR) signal plays key roles in the PCa progression, targeting androgens via the current androgen deprivation therapy (ADT) is the main therapeutic strategy for advanced PCa. However, most patients who receive ADT, including the second generation anti-androgens enzalutamide (also known as MDV3100) may finally develop the castration (or anti-androgen) resistance after 12-24 months treatment. In the manuscript by Asangani et al., the authors demonstrated that targeting the amino-terminal bromodomains of BRD4 could preferentially suppress human castration-resistant prostate cancer (CRPC) cell lines. While further studies are required to understand the full impact of their findings, the innovative approach provides a potential novel epigenetic approach for the concerted blockade of oncogenic drivers in CRPC.
Collapse
Affiliation(s)
- Si-Meng Wen
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chang-Yi Quan
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ning Jiang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Zhi-Qun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Yuan-Jie Niu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
85
|
Wen S, Chang HC, Tian J, Shang Z, Niu Y, Chang C. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:293-301. [PMID: 25432062 PMCID: PMC4305176 DOI: 10.1016/j.ajpath.2014.10.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 10/13/2014] [Accepted: 10/20/2014] [Indexed: 02/05/2023]
Abstract
The prostate is an androgen-sensitive organ that needs proper androgen/androgen receptor (AR) signals for normal development. The progression of prostate diseases, including benign prostate hyperplasia (BPH) and prostate cancer (PCa), also needs proper androgen/AR signals. Tissue recombination studies report that stromal, but not epithelial, AR plays more critical roles via the mesenchymal-epithelial interactions to influence the early process of prostate development. However, in BPH and PCa, much more attention has been focused on epithelial AR roles. However, accumulating evidence indicates that stromal AR is also irreplaceable and plays critical roles in prostate disease progression. Herein, we summarize the roles of stromal AR in the development of normal prostate, BPH, and PCa, with evidence from the recent results of in vitro cell line studies, tissue recombination experiments, and AR knockout animal models. Current evidence suggests that stromal AR may play positive roles to promote BPH and PCa progression, and targeting stromal AR selectively with AR degradation enhancer, ASC-J9, may allow development of better therapies with fewer adverse effects to battle BPH and PCa.
Collapse
Affiliation(s)
- Simeng Wen
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Hong-Chiang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Jing Tian
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York; Sex Hormone Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
86
|
Ding X, Yang DR, Xia L, Chen B, Yu S, Niu Y, Wang M, Li G, Chang C. Targeting TR4 nuclear receptor suppresses prostate cancer invasion via reduction of infiltrating macrophages with alteration of the TIMP-1/MMP2/MMP9 signals. Mol Cancer 2015; 14:16. [PMID: 25623427 PMCID: PMC4316804 DOI: 10.1186/s12943-014-0281-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 12/22/2014] [Indexed: 12/30/2022] Open
Abstract
Background TR4 nuclear receptor 4 (TR4) plays an important role in macrophages-associated foam cell formation of cardiovascular diseases and infiltrating macrophages are critical for prostate cancer (PCa) progression. However, the linkage of macrophages and TR4 and their impacts on PCa metastasis remains unclear. Results Knocking-down TR4 in human PCa cells (C4-2, CWR22Rv1), but not in human macrophages cells (THP-1), led to suppress the macrophages infiltration to PCa cells. The consequences of such suppression of the recruitment of macrophages toward PCa then resulted in suppressing the PCa cell invasion. Mechanism dissection found that knocking-down TR4 in PCa cells suppressed metastasis-related genes including MMP2, with induction of TIMP-1. Interruption assays using TIMP-1 neutralizing antibody could then reverse TR4-macrophage-mediated PCa invasion. IHC staining showed higher TR4 level, more macrophage infiltration, lower TIMP-1 and stronger MMP2/MMP9 in tumor tissues of the Gleason score 5 + 4 patients compared with the Gleason score 3 + 3 patients. Conclusion Targeting TR4 in prostate tumor microenvironment might represent a potential new therapeutic approach to better battle PCa metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12943-014-0281-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xianfan Ding
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China. .,George Whipple Lab for Cancer Research, Departments of Pathology, Urology and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14646, USA.
| | - Dong-Rong Yang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14646, USA. .,Department of Urology, the 2nd Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Liqun Xia
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Bide Chen
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Shicheng Yu
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China.
| | - Mingchao Wang
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Gonghui Li
- Department of Urology and Chawnshang Chang Liver Cancer Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14646, USA. .,Sex Hormone Research Center, China Medical University/Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
87
|
Shang Z, Cai Q, Zhang M, Zhu S, Ma Y, Sun L, Jiang N, Tian J, Niu X, Chen J, Sun Y, Niu Y. A switch from CD44⁺ cell to EMT cell drives the metastasis of prostate cancer. Oncotarget 2015; 6:1202-16. [PMID: 25483103 PMCID: PMC4359227 DOI: 10.18632/oncotarget.2841] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 11/24/2014] [Indexed: 01/10/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been linked to cancer stem-like (CD44+) cell in the prostate cancer (PCa) metastasis. However, the molecular mechanism remains elusive. Here, we found EMT contributed to metastasis in PCa patients failed in androgen deprivation therapy (ADT). Castration TRAMP model also proved PCa treated with ADT promoted EMT with increased CD44+ stem-like cells. Switched CD44+ cell to EMT cell is a key step for luminal PCa cell metastasis. Our results also suggested ADT might go through promoting TGFβ1-CD44 signaling to enhance swift to EMT. Targeting CD44 with salinomycin and siRNA could inhibit cell transition and decrease PCa invasion. Together, cancer stem-like (CD44+) cells could be the initiator cells of EMT modulated by TGFβ1-CD44 signaling. Combined therapy of ADT with anti-CD44 may become a new potential therapeutic approach to battle later stage PCa.
Collapse
Affiliation(s)
- Zhiqun Shang
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qiliang Cai
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Minghao Zhang
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shimiao Zhu
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuan Ma
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Libin Sun
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ning Jiang
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jing Tian
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaodan Niu
- University of Rochester, Rochester, New York, USA
| | - Jiatong Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai university, Tianjin, China
| | - Yinghao Sun
- Department of Urology, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - Yuanjie Niu
- Sex Hormone Research Center, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
88
|
|
89
|
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: 5.9] [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
|
90
|
Cao Z, Kyprianou N. WITHDRAWN: Mechanisms navigating the TGF-β pathway in prostate cancer. Asian J Urol 2014. [DOI: 10.1016/j.ajur.2014.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
91
|
Androgen receptor and immune inflammation in benign prostatic hyperplasia and prostate cancer. ACTA ACUST UNITED AC 2014; 4:935-950. [PMID: 26594314 DOI: 10.4155/cli.14.77] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Both benign prostatic hyperplasia (BPH) and prostate cancer (PCa) are frequent diseases in middle-aged to elderly men worldwide. While both diseases are linked to abnormal growth of the prostate, the epidemiological and pathological features of these two prostate diseases are different. BPH nodules typically arise from the transitional zone, and, in contrast, PCa arises from the peripheral zone. Androgen deprivation therapy alone may not be sufficient to cure these two prostatic diseases due to its undesirable side effects. The alteration of androgen receptor-mediated inflammatory signals from infiltrating immune cells and prostate stromal/epithelial cells may play key roles in those unwanted events. Herein, this review will focus on the roles of androgen/androgen receptor signals in the inflammation-induced progression of BPH and PCa.
Collapse
|
92
|
Helsen C, Van den Broeck T, Voet A, Prekovic S, Van Poppel H, Joniau S, Claessens F. Androgen receptor antagonists for prostate cancer therapy. Endocr Relat Cancer 2014; 21:T105-18. [PMID: 24639562 DOI: 10.1530/erc-13-0545] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Androgen deprivation is the mainstay therapy for metastatic prostate cancer (PCa). Another way of suppressing androgen receptor (AR) signaling is via AR antagonists or antiandrogens. Despite being frequently prescribed in clinical practice, there is conflicting evidence concerning the role of AR antagonists in the management of PCa. In the castration-resistant settings of PCa, docetaxel has been the only treatment option for decades. With recent evidence that castration-resistant PCa is far from AR-independent, there has been an increasing interest in developing new AR antagonists. This review gives a concise overview of the clinically available antiandrogens and the experimental AR antagonists that tackle androgen action with a different approach.
Collapse
Affiliation(s)
- Christine Helsen
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Thomas Van den Broeck
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, JapanLaboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Arnout Voet
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Stefan Prekovic
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Hendrik Van Poppel
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Steven Joniau
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Frank Claessens
- Laboratory of Molecular EndocrinologyDepartment of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-3000 Leuven, BelgiumUrologyDepartment of Development and Regeneration, University Hospitals Leuven, Herestraat 49, 3000 Leuven, BelgiumLaboratory for Structural BioinformaticsCenter for Life Science Technologies, RIKEN, Yokohama, Japan
| |
Collapse
|
93
|
Hu S, Li L, Yeh S, Cui Y, Li X, Chang HC, Jin J, Chang C. Infiltrating T cells promote prostate cancer metastasis via modulation of FGF11→miRNA-541→androgen receptor (AR)→MMP9 signaling. Mol Oncol 2014; 9:44-57. [PMID: 25135278 DOI: 10.1016/j.molonc.2014.07.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/31/2022] Open
Abstract
Early clinical studies suggested infiltrating T cells might be associated with poor outcomes in prostate cancer (PCa) patients. The detailed mechanisms how T cells contribute to PCa progression, however, remained unclear. Here, we found PCa cells have a better capacity to recruit more CD4(+) T cells than the surrounding normal prostate cells via secreting more chemokines-CXCL9. The consequences of more recruited CD4(+) T cells to PCa might then lead to enhance PCa cell invasion. Mechanism dissection revealed that infiltrating CD4(+) T cells might function through the modulation of FGF11→miRNA-541 signals to suppress PCa androgen receptor (AR) signals. The suppressed AR signals might then alter the MMP9 signals to promote the PCa cell invasion. Importantly, suppressed AR signals via AR-siRNA or anti-androgen Enzalutamide in PCa cells also enhanced the recruitment of T cells and the consequences of this positive feed back regulation could then enhance the PCa cell invasion. Targeting these newly identified signals via FGF11-siRNA, miRNA-541 inhibitor or MMP9 inhibitor all led to partially reverse the enhanced PCa cell invasion. Results from in vivo mouse models also confirmed the in vitro cell lines in co-culture studies. Together, these results concluded that infiltrating CD4(+) T cells could promote PCa metastasis via modulation of FGF11→miRNA-541→AR→MMP9 signaling. Targeting these newly identified signals may provide us a new potential therapeutic approach to better battle PCa metastasis.
Collapse
Affiliation(s)
- Shuai Hu
- Department of Urology, Peking University First Hospital, Beijing, China; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lei Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Yun Cui
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Xin Li
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Hong-Chiang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Jie Jin
- Department of Urology, Peking University First Hospital, Beijing, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA; Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan.
| |
Collapse
|
94
|
Ding X, Yang DR, Lee SO, Chen YL, Xia L, Lin SJ, Yu S, Niu YJ, Li G, Chang C. TR4 nuclear receptor promotes prostate cancer metastasis via upregulation of CCL2/CCR2 signaling. Int J Cancer 2014; 136:955-64. [PMID: 24975468 DOI: 10.1002/ijc.29049] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 06/13/2014] [Indexed: 11/05/2022]
Abstract
Testicular nuclear receptor 4 (TR4) plays protective roles against oxidative stress and DNA damage and might contribute to aging. Our recent clinical tumor tissue staining results showed higher expression of TR4 in prostate cancer (PCa) patients with high Gleason scores compared to the tissues with the low Gleason scores. In vitro migration/invasion assays after manipulation of the TR4 expression in PCa cells showed that TR4 promoted PCa cells migration/invasion. Mechanism dissection found that the CCL2/CCR2 signal plays the key role in the mediation of TR4-promoted PCa cells migration/invasion. Chromatin immunoprecipitation and Luciferase assays further confirmed TR4 modulation of CCL2 at the transcriptional level and addition of the CCR2 antagonist led to interruption of the TR4-enhanced PCa cells migration/invasion. Finally, the orthotopic xenografted mice studies using the luciferase expressing CWR22Rv1 cells found that TR4 enhanced PCa metastasis and this increased metastasis was reversed when the CCR2 antagonist was injected into the mice. Together, these in vitro and in vivo results revealed a positive role of TR4 in PCa metastasis and demonstrated CCL2/CCR2 signaling as an important mediator in exerting TR4 action. This finding suggests that TR4 may represent a biomarker related to PCa metastasis and targeting the TR4-CCL2/CCR2 axis may become a new therapeutic approach to battle PCa metastasis.
Collapse
Affiliation(s)
- Xianfan Ding
- Chawnshang Chang Liver Cancer Center, Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejang University, Hangzhou, China; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Gong Y, Chippada-Venkata UD, Oh WK. Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers (Basel) 2014; 6:1298-327. [PMID: 24978435 PMCID: PMC4190542 DOI: 10.3390/cancers6031298] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/31/2014] [Accepted: 06/09/2014] [Indexed: 01/16/2023] Open
Abstract
Matrix metalloproteinases (MMPs), a group of zinc-dependent endopeptidases involved in the degradation of the extracellular matrix, play an important role in tissue remodeling associated with various physiological processes such as morphogenesis, angiogenesis, and tissue repair, as well as pathological processes including cirrhosis, arthritis and cancer. The MMPs are well established as mediators of tumor invasion and metastasis by breaking down connective tissue barriers. Although there has been a vast amount of literature on the role of MMPs in invasion, metastasis and angiogenesis of various cancers, the role of these endopeptidases in prostate cancer progression has not been systematically reviewed. This overview summarizes findings on the tissue and blood expression of MMPs, their function, regulation and prognostic implication in human prostate cancer, with a focus on MMP-2, -7, -9, MT1-MMP and tissue inhibitor of metalloproteinase 1 (TIMP-1). This review also summarizes the efficacy and failure of early-generation matrix metalloproteinase inhibitors (MMPIs) in the treatment of metastatic prostate cancer and highlights the lessons and challenges for next generation MMPIs.
Collapse
Affiliation(s)
- Yixuan Gong
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Uma D Chippada-Venkata
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William K Oh
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
96
|
Abstract
INTRODUCTION The androgen receptor (AR) is a ligand-activated transcription factor that is expressed in primary and metastatic prostate cancers. There are advances in endocrine therapy for prostate cancer that are based on improved understanding of AR function. AREAS COVERED PubMed has been used to include most important publications on targeting the AR in prostate cancer. AR expression may be downregulated by agents used for chemoprevention of prostate cancer or, in models of advanced prostate cancer, by antisense oligonucleotides. New drugs that inhibit the steroidogenic enzyme CYP17A1 (abiraterone acetate) or diminish nuclear translocation of the AR (enzalutamide) have been shown to improve patients' survival in prostate cancer. However, it is clear that there is a development of resistance to these novel therapies. They may include increased expression of truncated, constitutively active AR or activation of the signaling pathway of signal transducers and activators of transcription. EXPERT OPINION Although introduction of novel drugs have improved patients' survival, there is a need to investigate the mechanisms of resistance further. The role of truncated AR and compensatory activation of signaling pathways as well as the development of scientifically justified combination therapies seems to be issues of a high priority.
Collapse
Affiliation(s)
- Zoran Culig
- Innsbruck Medical University, Experimental Urology, Department of Urology , Anichstrasse 35, A-6020 Innsbruck , Austria +43 512 504 24717 ; +43 512 504 24817 ;
| |
Collapse
|
97
|
Lin SJ, Zhang Y, Liu NC, Yang DR, Li G, Chang C. Minireview: Pathophysiological roles of the TR4 nuclear receptor: lessons learned from mice lacking TR4. Mol Endocrinol 2014; 28:805-21. [PMID: 24702179 DOI: 10.1210/me.2013-1422] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Testicular nuclear receptor 4 (TR4), also known as NR2C2, belongs to the nuclear receptor superfamily and shares high homology with the testicular nuclear receptor 2. The natural ligands of TR4 remained unclear until the recent discoveries of several energy/lipid sensors including the polyunsaturated fatty acid metabolites, 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, and their synthetic ligands, thiazolidinediones, used for treatment of diabetes. TR4 is widely expressed throughout the body and particularly concentrated in the testis, prostate, cerebellum, and hippocampus. It has been shown to play important roles in cerebellar development, forebrain myelination, folliculogenesis, gluconeogenesis, lipogenesis, muscle development, bone development, and prostate cancer progression. Here we provide a comprehensive summary of TR4 signaling including its upstream ligands/activators/suppressors, transcriptional coactivators/repressors, downstream targets, and their in vivo functions with potential impacts on TR4-related diseases. Importantly, TR4 shares similar ligands/activators with another key nuclear receptor, peroxisome proliferator-activated receptor γ, which raised several interesting questions about how these 2 nuclear receptors may collaborate with or counteract each other's function in their related diseases. Clear dissection of such molecular mechanisms and their differential roles in various diseases may help researchers to design new potential drugs with better efficacy and fewer side effects to battle TR4 and peroxisome proliferator-activated receptor γ involved diseases.
Collapse
Affiliation(s)
- Shin-Jen Lin
- George Whipple Laboratory for Cancer Research (S.-J.L., Y.Z., N.-C.L., C.C.), Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14646; Department of Urology (D.-R.Y.), the Second Affiliated Hospital of Suzhou University, Suzhou, 215004 China; Chawnshang Chang Liver Cancer Center and Department of Urology (G.L.), Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016 China; and Sex Hormone Research Center (C.C.), China Medical University/Hospital, Taichung, 404 Taiwan
| | | | | | | | | | | |
Collapse
|
98
|
Shiota M, Yokomizo A, Takeuchi A, Imada K, Kashiwagi E, Song Y, Inokuchi J, Tatsugami K, Uchiumi T, Naito S. Inhibition of protein kinase C/Twist1 signaling augments anticancer effects of androgen deprivation and enzalutamide in prostate cancer. Clin Cancer Res 2013; 20:951-61. [PMID: 24352647 DOI: 10.1158/1078-0432.ccr-13-1809] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The progression of prostate cancer to metastatic and castration-resistant disease represents a critical step. We previously showed that the transcription factor Twist1, which promotes epithelial-mesenchymal transition, was involved in castration-resistant progression. Similarly, protein kinase C (PKC) has been implicated in both metastatic progression and castration resistance in prostate cancer. EXPERIMENTAL DESIGN In this study, we aimed to elucidate the role of PKC/Twist1 signaling in castration resistance, and to apply this information to the development of a novel therapeutic concept using PKC inhibitor Ro31-8220 against prostate cancer using various prostate cancer cell lines. RESULTS Androgen deprivation and the next-generation antiandrogen enzalutamide induced PKC activation and Twist1 expression, which were reversed by the PKC inhibitor Ro31-8220. Ro31-8220 suppressed cell proliferation in androgen-dependent prostate cancer LNCaP cells, which was augmented by its combination with androgen deprivation or enzalutamide. The favorable anticancer effects of the combination of Ro31-8220 and enzalutamide were also observed in castration-resistant C4-2 and 22Rv1 cells. Furthermore, PKC phosphorylation was elevated in castration-resistant and enzalutamide-resistant cells compared with their parental cells, leading to persistent sensitivity to Ro-31-8220 in castration- and enzalutamide-resistant cells. CONCLUSIONS Taken together, these findings indicate that PKC/Twist1 signaling contributes to castration resistance as well as enzalutamide resistance in prostate cancer, and suggest that therapeutics targeting PKC/Twist1 signaling, such as PKC inhibitors, represent a promising novel therapeutic strategy for prostate cancer, especially castration-resistant prostate cancer, when combined with enzalutamide.
Collapse
Affiliation(s)
- Masaki Shiota
- Authors' Affiliations: Departments of Urology and Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Sun F, Chen HG, Li W, Yang X, Wang X, Jiang R, Guo Z, Chen H, Huang J, Borowsky AD, Qiu Y. Androgen receptor splice variant AR3 promotes prostate cancer via modulating expression of autocrine/paracrine factors. J Biol Chem 2013; 289:1529-39. [PMID: 24297183 DOI: 10.1074/jbc.m113.492140] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Deregulation of androgen receptor (AR) splice variants has been implicated to play a role in prostate cancer development and progression. To understand their functions in prostate, we established a transgenic mouse model (AR3Tg) with targeted expression of the constitutively active and androgen-independent AR splice variant AR3 (a.k.a. AR-V7) in prostate epithelium. We found that overexpression of AR3 modulates expression of a number of tumor-promoting autocrine/paracrine growth factors (including Tgfβ2 and Igf1) and expands prostatic progenitor cell population, leading to development of prostatic intraepithelial neoplasia. In addition, we showed that some epithelial-mesenchymal transition-associated genes are up-regulated in AR3Tg prostates, suggesting that AR3 may antagonize AR activity and halt the differentiation process driven by AR and androgen. This notion is supported by our observations that the number of Ck5(+)/Ck8(+) intermediate cells is increased in AR3Tg prostates after castration, and expression of AR3 transgene in these intermediate cells compromises prostate epithelium regeneration upon androgen replacement. Our results demonstrate that AR3 is a driver of prostate cancer, at least in part, through modulating multiple tumor-promoting autocrine/paracrine factors.
Collapse
Affiliation(s)
- Feng Sun
- From the Departments of Pharmacology and The Greenebaum Cancer Center, and
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Soh SF, Huang CK, Lee SO, Xu D, Yeh S, Li J, Yong EL, Gong Y, Chang C. Determination of androgen receptor degradation enhancer ASC-J9(®) in mouse sera and organs with liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal 2013; 88:117-22. [PMID: 24042123 DOI: 10.1016/j.jpba.2013.08.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 08/18/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022]
Abstract
A novel androgen receptor (AR) degradation enhancer ASC-J9(®) has displayed beneficial effects during the in vitro and in vivo studies for treatment of prostate cancer, liver cancer, bladder cancer and spinal and bulbar muscular atrophy (SBMA). It works mainly via the degradation of AR with minimal side effects on the tested mice. Here we developed a fast, robust and more sensitive method for the quantification of ASC-J9(®) in 100μL of mouse serum by using liquid chromatography tandem mass spectrometry (LC-MS/MS). The limit of quantification (LOQ) was found to be 5nM for ASCJ9(®). This method was successfully applied to investigate the pharmacokinetics of ASC-J9(®) in mice serum samples and also the distribution of the drug in various mice organs after single dose injection with results showing that ASC-J9(®) could be quickly absorbed in vivo and had a relatively slow elimination half-life of 5.45h. The ASC-J9(®) also exhibited a higher tendency to accumulate in organs such as liver, testes and prostate.
Collapse
Affiliation(s)
- Shu Fang Soh
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Lower Kent Ridge Road, Singapore 119074; George Whipple Lab for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|