1
|
Xie J, Yang MR, Hu X, Hong ZS, Bai YY, Sheng J, Tian Y, Shi CY. Moringa oleifera Lam. Isothiocyanate Quinazolinone Derivatives Inhibit U251 Glioma Cell Proliferation through Cell Cycle Regulation and Apoptosis Induction. Int J Mol Sci 2023; 24:11376. [PMID: 37511135 PMCID: PMC10379366 DOI: 10.3390/ijms241411376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
A major active constituent of Moringa oleifera Lam. is 4-[(α-L-rhamnose oxy) benzyl] isothiocyanate (MITC). To broaden MITC's application and improve its biological activity, we synthesized a series of MITC quinazolinone derivatives and evaluated their anticancer activity. The anticancer effects and mechanisms of the compound with the most potent anticancer activity were investigated further. Among 16 MITC quinazolinone derivatives which were analyzed, MITC-12 significantly inhibited the growth of U251, A375, A431, HCT-116, HeLa, and MDA-MB-231 cells. MITC-12 significantly inhibited U251 cell proliferation in a time- and dose-dependent manner and decreased the number of EdU-positive cells, but was not toxic to normal human gastric mucosal cells (GES-1). Further, MITC-12 induced apoptosis of U251 cells, and increased caspase-3 expression levels and the Bax:Bcl-2 ratio. In addition, MITC-12 significantly decreased the proportion of U251 cells in the G1 phase and increased it in S and G2 phases. Transcriptome sequencing showed that MITC-12 had a significant regulatory effect on pathways regulating the cell cycle. Further, MITC-12 significantly decreased the expression levels of the cell cycle-related proteins CDK2, cyclinD1, and cyclinE, and increased those of cyclinA2, as well as the p-JNK:JNK ratio. These results indicate that MITC-12 inhibits U251 cell proliferation by inducing apoptosis and cell cycle arrest, activating JNK, and regulating cell cycle-associated proteins. MITC-12 has potential for use in the prevention and treatment of glioma.
Collapse
Affiliation(s)
- Jing Xie
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Ming-Rong Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Rural Science and Technology Service Center, Kunming 650021, China
| | - Xia Hu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Zi-Shan Hong
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Yu-Ying Bai
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming 650201, China
| | - Jun Sheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yang Tian
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Chong-Ying Shi
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
2
|
Kumar R, Sena LA, Denmeade SR, Kachhap S. The testosterone paradox of advanced prostate cancer: mechanistic insights and clinical implications. Nat Rev Urol 2023; 20:265-278. [PMID: 36543976 PMCID: PMC10164147 DOI: 10.1038/s41585-022-00686-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
The discovery of the benefits of castration for prostate cancer treatment in 1941 led to androgen deprivation therapy, which remains a mainstay of the treatment of men with advanced prostate cancer. However, as early as this original publication, the inevitable development of castration-resistant prostate cancer was recognized. Resistance first manifests as a sustained rise in the androgen-responsive gene, PSA, consistent with reactivation of the androgen receptor axis. Evaluation of clinical specimens demonstrates that castration-resistant prostate cancer cells remain addicted to androgen signalling and adapt to chronic low-testosterone states. Paradoxically, results of several studies have suggested that treatment with supraphysiological levels of testosterone can retard prostate cancer growth. Insights from these studies have been used to investigate administration of supraphysiological testosterone to patients with prostate cancer for clinical benefits, a strategy that is termed bipolar androgen therapy (BAT). BAT involves rapid cycling from supraphysiological back to near-castration testosterone levels over a 4-week cycle. Understanding how BAT works at the molecular and cellular levels might help to rationalize combining BAT with other agents to achieve increased efficacy and tumour responses.
Collapse
Affiliation(s)
- Rajendra Kumar
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Laura A Sena
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Sushant Kachhap
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
3
|
Gupta A, Carnazza M, Jones M, Darzynkiewicz Z, Halicka D, O'Connell T, Zhao H, Dadafarin S, Shin E, Schwarcz MD, Moscatello A, Tiwari RK, Geliebter J. Androgen Receptor Activation Induces Senescence in Thyroid Cancer Cells. Cancers (Basel) 2023; 15:cancers15082198. [PMID: 37190127 DOI: 10.3390/cancers15082198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy, with an approximately three-fold higher incidence in women. TCGA data indicate that androgen receptor (AR) RNA is significantly downregulated in PTC. In this study, AR-expressing 8505C (anaplastic TC) (84E7) and K1 (papillary TC) cells experienced an 80% decrease in proliferation over 6 days of exposure to physiological levels of 5α-dihydrotestosterone (DHT). In 84E7, continuous AR activation resulted in G1 growth arrest, accompanied by a flattened, vacuolized cell morphology, with enlargement of the cell and the nuclear area, which is indicative of senescence; this was substantiated by an increase in senescence-associated β-galactosidase activity, total RNA and protein content, and reactive oxygen species. Additionally, the expression of tumor suppressor proteins p16, p21, and p27 was significantly increased. A non-inflammatory senescence-associated secretory profile was induced, significantly decreasing inflammatory cytokines and chemokines such as IL-6, IL-8, TNF, RANTES, and MCP-1; this is consistent with the lower incidence of thyroid inflammation and cancer in men. Migration increased six-fold, which is consistent with the clinical observation of increased lymph node metastasis in men. Proteolytic invasion potential was not significantly altered, which is consistent with unchanged MMP/TIMP expression. Our studies provide evidence that the induction of senescence is a novel function of AR activation in thyroid cancer cells, and may underlie the protective role of AR activation in the decreased incidence of TC in men.
Collapse
Affiliation(s)
- Anvita Gupta
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Michelle Carnazza
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Melanie Jones
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Zbigniew Darzynkiewicz
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA
| | - Dorota Halicka
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Timmy O'Connell
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Hong Zhao
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Sina Dadafarin
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
| | - Edward Shin
- Department of Otolaryngology, New York Eye and Ear Infirmary of Mount Sinai, New York, NY 10003, USA
| | - Monica D Schwarcz
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Raj K Tiwari
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Jan Geliebter
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
4
|
Shin SY, Centenera MM, Hodgson JT, Nguyen EV, Butler LM, Daly RJ, Nguyen LK. A Boolean-based machine learning framework identifies predictive biomarkers of HSP90-targeted therapy response in prostate cancer. Front Mol Biosci 2023; 10:1094321. [PMID: 36743211 PMCID: PMC9892654 DOI: 10.3389/fmolb.2023.1094321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Precision medicine has emerged as an important paradigm in oncology, driven by the significant heterogeneity of individual patients' tumour. A key prerequisite for effective implementation of precision oncology is the development of companion biomarkers that can predict response to anti-cancer therapies and guide patient selection for clinical trials and/or treatment. However, reliable predictive biomarkers are currently lacking for many anti-cancer therapies, hampering their clinical application. Here, we developed a novel machine learning-based framework to derive predictive multi-gene biomarker panels and associated expression signatures that accurately predict cancer drug sensitivity. We demonstrated the power of the approach by applying it to identify response biomarker panels for an Hsp90-based therapy in prostate cancer, using proteomic data profiled from prostate cancer patient-derived explants. Our approach employs a rational feature section strategy to maximise model performance, and innovatively utilizes Boolean algebra methods to derive specific expression signatures of the marker proteins. Given suitable data for model training, the approach is also applicable to other cancer drug agents in different tumour settings.
Collapse
Affiliation(s)
- Sung-Young Shin
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Margaret M. Centenera
- South Australian Immunogenomics Cancer Institute and Freemasons Foundation Centre for Men’s Health, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Joshua T. Hodgson
- South Australian Immunogenomics Cancer Institute and Freemasons Foundation Centre for Men’s Health, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Elizabeth V. Nguyen
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lisa M. Butler
- South Australian Immunogenomics Cancer Institute and Freemasons Foundation Centre for Men’s Health, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Roger J. Daly
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lan K. Nguyen
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
5
|
Xie Q, Fan X, Han Y, Wu BX, Zhu B. Daphnoretin Arrests the Cell Cycle and Induces Apoptosis in Human Breast Cancer Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:2332-2339. [PMID: 36154031 DOI: 10.1021/acs.jnatprod.2c00504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Emerging evidence has shown that daphnoretin, one of the main active ingredients of Daphne giraldii Nitsche, processes antitumor activities in several tumor cells (e.g., colon cancer, lung cancer, cervical cancer, and osteosarcoma). However, the antitumor effect and its mechanism in breast cancer are unexplored. In this study, our data indicated that daphnoretin obviously suppressed the proliferation of breast cancer MCF-7 and MDA-MB-231 cells. Further studies showed that daphnoretin remarkably increased the p21 level, decreased cyclin E and CDK2 levels, and then arrested the cell cycle at the S phase. Moreover, daphnoretin obviously lowered the BCL-2 level and raised the levels of BAX and cleaved caspase-9 and -3, leading to cell apoptosis. Furthermore, daphnoretin remarkably decreased the ratio of p-PI3K/PI3K and p-AKT/AKT in breast cancer cells. Collectively, these findings demonstrated that daphnoretin could suppress breast cancer cell proliferation through cell cycle arrest and inducing apoptosis, which is related to the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qi Xie
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu China
| | - Xinmei Fan
- Jiangsu Food & Pharmaceutical Science College, 4 Meicheng Road, Huaian, 223003, Jiangsu People's Republic of China
| | - Yonghong Han
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu People's Republic of China
| | - Bao-Xiang Wu
- Key Laboratory of Coal Processing and Efficient Utilization, Ministry of Education, China University of Mining & Technology, Xuzhou221116, Jiangsu People's Republic of China
| | - Bao Zhu
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu People's Republic of China
| |
Collapse
|
6
|
Chen L, Sun Y, Tang M, Wu D, Xiang Z, Huang CP, You B, Xie D, Ye Q, Yu D, Chang C. High-dose-androgen-induced autophagic cell death to suppress the Enzalutamide-resistant prostate cancer growth via altering the circRNA-BCL2/miRNA-198/AMBRA1 signaling. Cell Death Dis 2022; 8:128. [PMID: 35318303 PMCID: PMC8941094 DOI: 10.1038/s41420-022-00898-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 01/22/2023]
Abstract
Androgen deprivation therapy (ADT) is a gold standard treatment for advanced PCa. However, most patients eventually develop the castration-resistant prostate cancer (CRPC) that progresses rapidly despite ongoing systemic androgen deprivation. While early studies indicated that high physiological doses of androgens might suppress rather than promote PCa cell growth in some selective CRPC patients, the exact mechanism of this opposite effect remains unclear. Here we found that Enzalutamide-resistant (EnzR) CRPC cells can be suppressed by the high-dose-androgen (dihydrotestosterone, DHT). Mechanism dissection suggested that a high-dose-DHT can suppress the circular RNA-BCL2 (circRNA-BCL2) expression via transcriptional regulation of its host gene BCL2. The suppressed circRNA-BCL2 can then alter the expression of miRNA-198 to modulate the AMBRA1 expression via direct binding to the 3′UTR of AMBRA1 mRNA. The consequences of high-dose-DHT suppressed circRNA-BCL2/miRNA-198/AMBRA1 signaling likely result in induction of the autophagic cell death to suppress the EnzR CRPC cell growth. Preclinical studies using in vivo xenograft mouse models also demonstrated that AMBRA1-shRNA to suppress the autophagic cell death can weaken the effect of high-dose-DHT on EnzR CRPC tumors. Together, these in vitro and in vivo data provide new insights for understanding the mechanisms underlying high-dose-DHT suppression of the EnzR CRPC cell growth, supporting a potential therapy using high-dose-androgens to suppress CRPC progression in the future.
Collapse
Affiliation(s)
- Lei Chen
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China.,George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Min Tang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA.,Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji Universiry, Shanghai, 200092, China
| | - Zhendong Xiang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji Universiry, Shanghai, 200092, China
| | - Chi-Ping Huang
- Sex Hormone Research Center, Department of Urology, China Medical University/Hospital, Taichung, 404, Taiwan, ROC
| | - Bosen You
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Dongdong Xie
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China
| | - Qinglin Ye
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China
| | - Dexin Yu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA. .,Sex Hormone Research Center, Department of Urology, China Medical University/Hospital, Taichung, 404, Taiwan, ROC.
| |
Collapse
|
7
|
Cell cycle involvement in cancer therapy; WEE1 kinase, a potential target as therapeutic strategy. Mutat Res 2022; 824:111776. [PMID: 35247630 DOI: 10.1016/j.mrfmmm.2022.111776] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/22/2022]
Abstract
Mitosis is the process of cell division and is regulated by checkpoints in the cell cycle. G1-S, S, and G2-M are the three main checkpoints that prevent initiation of the next phase of the cell cycle phase until previous phase has completed. DNA damage leads to activation of the G2-M checkpoint, which can trigger a downstream DNA damage response (DDR) pathway to induce cell cycle arrest while the damage is repaired. If the DNA damage cannot be repaired, the replication stress response (RSR) pathway finally leads to cell death by apoptosis, in this case called mitotic catastrophe. Many cancer treatments (chemotherapy and radiotherapy) cause DNA damages based on SSBs (single strand breaks) or DSBs (double strand breaks), which cause cell death through mitotic catastrophe. However, damaged cells can activate WEE1 kinase (as a part of the DDR and RSR pathways), which prevents apoptosis and cell death by inducing cell cycle arrest at G2 phase. Therefore, inhibition of WEE1 kinase could sensitize cancer cells to chemotherapeutic drugs. This review focuses on the role of WEE1 kinase (as a biological macromolecule which has a molecular mass of 96 kDa) in the cell cycle, and its interactions with other regulatory pathways. In addition, we discuss the potential of WEE1 inhibition as a new therapeutic approach in the treatment of various cancers, such as melanoma, breast cancer, pancreatic cancer, cervical cancer, etc.
Collapse
|
8
|
Nakayama H, Sekine Y, Oka D, Miyazawa Y, Arai S, Koike H, Matsui H, Shibata Y, Suzuki K. Combination therapy with novel androgen receptor antagonists and statin for castration-resistant prostate cancer. Prostate 2022; 82:314-322. [PMID: 34843630 DOI: 10.1002/pros.24274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/19/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND One of the growth mechanisms of castration-resistant prostate cancer (CRPC) is de novo androgen synthesis from intracellular cholesterol, and statins may be able to inhibit this mechanism. In addition, statins have been reported to suppress the expression of androgen receptors (ARs) in prostate cancer cell lines. In this study, we investigated a combination therapy of novel AR antagonists and statin, simvastatin, for CRPC. METHODS LNCaP, 22Rv1, and PC-3 human prostate cancer cell lines were used. We developed androgen-independent LNCaP cells (LNCaP-LA). Microarray analysis was performed, followed by pathway analysis, and mRNA and protein expression was evaluated by quantitative real-time polymerase chain reaction and Western blot analysis, respectively. Cell viability was determined by MTS assay and cell counts. All evaluations were performed on cells treated with simvastatin and with or without AR antagonists (enzalutamide, apalutamide, and darolutamide). RESULTS The combination of darolutamide and simvastatin most significantly suppressed proliferation in LNCaP-LA and 22Rv1 cells. In a 22Rv1-derived mouse xenograft model, the combination of darolutamide and simvastatin enhanced the inhibition of cell proliferation. In LNCaP-LA cells, the combination of darolutamide and simvastatin led to reduction in the mRNA expression of the androgen-stimulated genes, KLK2 and PSA; however, this reduction in expression did not occur in 22Rv1 cells. The microarray data and pathway analyses showed that the number of differentially expressed genes in the darolutamide and simvastatin-treated 22Rv1 cells was the highest in the pathway termed "role of cell cycle." Consequently, we focused our efforts on the cell cycle regulator polo-like kinase 1 (PLK1), cyclin-dependent kinase 2 (CDK2), and cell cycle division 25C (CDC25C). In 22Rv1 cells, the combination of darolutamide and simvastatin suppressed the mRNA and protein expression of these three genes. In addition, in PC-3 cells (which lack AR expression), the combination of simvastatin and darolutamide enhanced the suppression of cell proliferation and expression of these genes. CONCLUSIONS Simvastatin alters the expression of many genes involved in the cell cycle in CRPC cells. Thus, the combination of novel AR antagonists (darolutamide) and simvastatin can potentially affect CRPC growth through both androgen-dependent and androgen-independent mechanisms.
Collapse
Affiliation(s)
- Hiroshi Nakayama
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Daisuke Oka
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshiyuki Miyazawa
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Seiji Arai
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hidekazu Koike
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroshi Matsui
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yasuhiro Shibata
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
9
|
Discovery proteomics defines androgen-regulated glycoprotein networks in prostate cancer cells, as well as putative biomarkers of prostatic diseases. Sci Rep 2021; 11:22208. [PMID: 34782677 PMCID: PMC8592995 DOI: 10.1038/s41598-021-01554-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/20/2021] [Indexed: 12/05/2022] Open
Abstract
Supraphysiologic androgen (SPA) inhibits cell proliferation in prostate cancer (PCa) cells by transcriptional repression of DNA replication and cell-cycle genes. In this study, quantitative glycoprotein profiling identified androgen-regulated glycoprotein networks associated with SPA-mediated inhibition of PCa cell proliferation, and androgen-regulated glycoproteins in clinical prostate tissues. SPA-regulated glycoprotein networks were enriched for translation factors and ribosomal proteins, proteins that are known to be O-GlcNAcylated in response to various cellular stresses. Thus, androgen-regulated glycoproteins are likely to be targeted for O-GlcNAcylation. Comparative analysis of glycosylated proteins in PCa cells and clinical prostate tissue identified androgen-regulated glycoproteins that are differentially expressed prostate tissues at various stages of cancer. Notably, the enzyme ectonucleoside triphosphate diphosphohydrolase 5 was found to be an androgen-regulated glycoprotein in PCa cells, with higher expression in cancerous versus non-cancerous prostate tissue. Our glycoproteomics study provides an experimental framework for characterizing androgen-regulated proteins and glycoprotein networks, toward better understanding how this subproteome leads to physiologic and supraphysiologic proliferation responses in PCa cells, and their potential use as druggable biomarkers of dysregulated AR-dependent signaling in PCa cells.
Collapse
|
10
|
Nordeen SK, Su LJ, Osborne GA, Hayman PM, Orlicky DJ, Wessells VM, van Bokhoven A, Flaig TW. Titration of Androgen Signaling: How Basic Studies Have Informed Clinical Trials Using High-Dose Testosterone Therapy in Castrate-Resistant Prostate Cancer. Life (Basel) 2021; 11:884. [PMID: 34575033 PMCID: PMC8465783 DOI: 10.3390/life11090884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Since the Nobel Prize-winning work of Huggins, androgen ablation has been a mainstay for treatment of recurrent prostate cancer. While initially effective for most patients, prostate cancers inevitably develop the ability to survive, grow, and metastasize further, despite ongoing androgen suppression. Here, we briefly review key preclinical studies over decades and include illustrative examples from our own laboratories that suggest prostate cancer cells titrate androgen signaling to optimize growth. Such laboratory-based studies argue that adaptations that allow growth in a low-androgen environment render prostate cancer sensitive to restoration of androgens, especially at supraphysiologic doses. Based on preclinical data as well as clinical observations, trials employing high-dose testosterone (HDT) therapy have now been conducted. These trials suggest a clinical benefit in cancer response and quality of life in a subset of castration-resistant prostate cancer patients. Laboratory studies also suggest that HDT may yet be optimized further to improve efficacy or durability of response. However, laboratory observations suggest that the cancer will inevitably adapt to HDT, and, as with prior androgen deprivation, disease progression follows. Nonetheless, the adaptations made to render tumors resistant to hormonal manipulations may reveal vulnerabilities that can be exploited to prolong survival and provide other clinical benefits.
Collapse
Affiliation(s)
- Steven K. Nordeen
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (S.K.N.); (D.J.O.); (A.v.B.)
| | - Lih-Jen Su
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (G.A.O.); (P.M.H.); (V.M.W.)
| | - Gregory A. Osborne
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (G.A.O.); (P.M.H.); (V.M.W.)
| | - Perry M. Hayman
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (G.A.O.); (P.M.H.); (V.M.W.)
| | - David J. Orlicky
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (S.K.N.); (D.J.O.); (A.v.B.)
| | - Veronica M. Wessells
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (G.A.O.); (P.M.H.); (V.M.W.)
| | - Adrie van Bokhoven
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (S.K.N.); (D.J.O.); (A.v.B.)
| | - Thomas W. Flaig
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (G.A.O.); (P.M.H.); (V.M.W.)
| |
Collapse
|
11
|
Kokal M, Mirzakhani K, Pungsrinont T, Baniahmad A. Mechanisms of Androgen Receptor Agonist- and Antagonist-Mediated Cellular Senescence in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12071833. [PMID: 32650419 PMCID: PMC7408918 DOI: 10.3390/cancers12071833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
The androgen receptor (AR) plays a leading role in the control of prostate cancer (PCa) growth. Interestingly, structurally different AR antagonists with distinct mechanisms of antagonism induce cell senescence, a mechanism that inhibits cell cycle progression, and thus seems to be a key cellular response for the treatment of PCa. Surprisingly, while physiological levels of androgens promote growth, supraphysiological androgen levels (SAL) inhibit PCa growth in an AR-dependent manner by inducing cell senescence in cancer cells. Thus, oppositional acting ligands, AR antagonists, and agonists are able to induce cellular senescence in PCa cells, as shown in cell culture model as well as ex vivo in patient tumor samples. This suggests a dual AR-signaling dependent on androgen levels that leads to the paradox of the rational to keep the AR constantly inactivated in order to treat PCa. These observations however opened the option to treat PCa patients with AR antagonists and/or with androgens at supraphysiological levels. The latter is currently used in clinical trials in so-called bipolar androgen therapy (BAT). Notably, cellular senescence is induced by AR antagonists or agonist in both androgen-dependent and castration-resistant PCa (CRPC). Pathway analysis suggests a crosstalk between AR and the non-receptor tyrosine kinase Src-Akt/PKB and the PI3K-mTOR-autophagy signaling in mediating AR-induced cellular senescence in PCa. In this review, we summarize the current knowledge of therapeutic induction and intracellular pathways of AR-mediated cellular senescence.
Collapse
Affiliation(s)
| | | | | | - Aria Baniahmad
- Correspondence: ; Tel.: +49-3641-9396820; Fax: +49-3641-99396822
| |
Collapse
|
12
|
Abstract
Introduction: The use of testosterone therapy (TTh) in men with prostate cancer (PCa) is relatively new, and controversial, due to the longstanding maxim that TTh is contraindicated in men with PCa. Scientific advances have prompted a reevaluation of the potential role for TTh in men with PCa, particularly as TTh has been shown to provide important symptomatic and general health benefits to men with testosterone deficiency (TD), including many men with PCa who may expect to live 30-50 years after diagnosis. Areas covered: This review outlines the historical underpinnings of the historical belief that TTh 'fuels' PCa and the experimental and clinical studies that have radically altered this view, including description of the saturation model. The authors review studies of TTh in men with PCa following radical prostatectomy and radiation therapy, in men on active surveillance, and in men with advanced or metastatic PCa. Expert opinion: TTh provides important symptomatic and overall health benefits for men with PCa who have TD. Although more safety studies are needed, TTh is a reasonable therapeutic option for men with low-risk PCa after surgery or radiation. Data in men on active surveillance are limited, but initial reports are reassuring.
Collapse
Affiliation(s)
- Abraham Morgentaler
- Beth Israel Deaconess Medical Center, Harvard Medical School, Men's Health Boston , Boston , MA , USA
| | | |
Collapse
|
13
|
Chatterjee P, Schweizer MT, Lucas JM, Coleman I, Nyquist MD, Frank SB, Tharakan R, Mostaghel E, Luo J, Pritchard CC, Lam HM, Corey E, Antonarakis ES, Denmeade SR, Nelson PS. Supraphysiological androgens suppress prostate cancer growth through androgen receptor-mediated DNA damage. J Clin Invest 2019; 129:4245-4260. [PMID: 31310591 PMCID: PMC6763228 DOI: 10.1172/jci127613] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/11/2019] [Indexed: 12/30/2022] Open
Abstract
Prostate cancer (PC) is initially dependent on androgen receptor (AR) signaling for survival and growth. Therapeutics designed to suppress AR activity serve as the primary intervention for advanced disease. However, supraphysiological androgen (SPA) concentrations can produce paradoxical responses leading to PC growth inhibition. We sought to discern the mechanisms by which SPA inhibits PC and to determine if molecular context associates with anti-tumor activity. SPA produced an AR-mediated, dose-dependent induction of DNA double-strand breaks (DSBs), G0/G1 cell cycle arrest and cellular senescence. SPA repressed genes involved in DNA repair and delayed the restoration of damaged DNA which was augmented by PARP1 inhibition. SPA-induced DSBs were accentuated in BRCA2-deficient PCs, and combining SPA with PARP or DNA-PKcs inhibition further repressed growth. Next-generation sequencing was performed on biospecimens from PC patients receiving SPA as part of ongoing Phase II clinical trials. Patients with mutations in genes mediating homology-directed DNA repair were more likely to exhibit clinical responses to SPA. These results provide a mechanistic rationale for directing SPA therapy to PCs with AR amplification or DNA repair deficiency, and for combining SPA therapy with PARP inhibition.
Collapse
Affiliation(s)
| | - Michael T. Schweizer
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | - Elahe Mostaghel
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jun Luo
- Department of Urology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Emmanuel S. Antonarakis
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Samuel R. Denmeade
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter S. Nelson
- Division of Human Biology and
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
Said R, Garcia-Mayea Y, Trabelsi N, Setti Boubaker N, Mir C, Blel A, Ati N, Paciucci R, Hernández-Losa J, Rammeh S, Derouiche A, Chebil M, LLeonart ME, Ouerhani S. Expression patterns and bioinformatic analysis of miR-1260a and miR-1274a in Prostate Cancer Tunisian patients. Mol Biol Rep 2018; 45:2345-2358. [PMID: 30250996 DOI: 10.1007/s11033-018-4399-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Abstract
Currently, microRNAs (miRs) represent great biomarkers in cancer due to their stability and their potential role in diagnosis, prognosis and therapy. This study aims to evaluate the expression levels of miRs-1260 and -1274a in prostate cancer (PC) samples and to identify their eventual targets by using bioinformatic analysis. In this project, we evaluated the expression status of miRs-1260 and -1274a in 86 PC patients and 19 controls by using real-time quantitative PCR and 2-ΔΔCt method. Moreover, we retrieved validated and predicted targets of miRs from several datasets by using the "multiMir" R/Bioconductor package. We have found that miRs-1260 and -1274a were over-expressed in PC patients compared to controls (p < 1 × 10-5). Moreover ROC curve for miRs-1260 and 1274a showed a good performance to distinguish between controls group and PC samples with an area under the ROC curve of 0.897 and 0.784 respectively. However, no significant association could be shown between these two miRs and clinical parameters such as PSA levels, Gleason score, tumor stage, D'Amico classification, lymph node metastasis statues, tumor recurrence, metastasis status and progression after a minimum of 5 years follow-up. Finally, a bioinformatic analysis revealed the association between these two miRs and several targets implicated in prostate cancer initiation pathways.
Collapse
Affiliation(s)
- Rahma Said
- Laboratory of Protein Engineering and Bio-active Molecules, National Institute of Applied Science and Technology - University of Carthage, Tunis, Tunisia
- Faculty of Sciences of Bizerte, University of Carthage, Tunis, Tunisia
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Vall d'Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035, Barcelona, Spain
| | - Nesrine Trabelsi
- Laboratory of Protein Engineering and Bio-active Molecules, National Institute of Applied Science and Technology - University of Carthage, Tunis, Tunisia
| | - Nouha Setti Boubaker
- Laboratory of Protein Engineering and Bio-active Molecules, National Institute of Applied Science and Technology - University of Carthage, Tunis, Tunisia
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Vall d'Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035, Barcelona, Spain
| | - Ahlem Blel
- Pathology Anatomy and Cytology Department, Charles Nicolle Hospital, Tunis, Tunisia
| | - Nidhal Ati
- Urology Department, Charles Nicolle Hospital, Tunis, Tunisia
| | - Rosanna Paciucci
- Biomedical Research Group of Urology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Hernández-Losa
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Vall d'Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035, Barcelona, Spain
| | - Soumaya Rammeh
- Pathology Anatomy and Cytology Department, Charles Nicolle Hospital, Tunis, Tunisia
| | - Amine Derouiche
- Urology Department, Charles Nicolle Hospital, Tunis, Tunisia
| | - Mohamed Chebil
- Urology Department, Charles Nicolle Hospital, Tunis, Tunisia
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Pathology Department, Vall d'Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035, Barcelona, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| | - Slah Ouerhani
- Laboratory of Protein Engineering and Bio-active Molecules, National Institute of Applied Science and Technology - University of Carthage, Tunis, Tunisia.
| |
Collapse
|
15
|
Isaacsson Velho P, Carducci MA. Investigational therapies targeting the androgen signaling axis and the androgen receptor and in prostate cancer – recent developments and future directions. Expert Opin Investig Drugs 2018; 27:811-822. [DOI: 10.1080/13543784.2018.1513490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
| | - Michael A. Carducci
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
- Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Wang K, Ruan H, Xu T, Liu L, Liu D, Yang H, Zhang X, Chen K. Recent advances on the progressive mechanism and therapy in castration-resistant prostate cancer. Onco Targets Ther 2018; 11:3167-3178. [PMID: 29881290 PMCID: PMC5983013 DOI: 10.2147/ott.s159777] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background Although there have been great advances in mechanisms and therapeutic methods of prostate cancer, the mortality rate of prostate cancer remains high. The castration-resistant prostate cancer (CRPC), which develops from hormone-sensitive prostate cancer, foreshadows a more dismal outcome. Concomitant with the researches in the mechanism of CRPC and therapy for CRPC, more and more landmark progress has been made in recent years. Methods A number of clinical and experimental studies were reviewed to indicate the novel advancement in the progressive mechanism and therapy of CRPC. Results The androgen receptor (AR) is still a vital driver in the progression of CRPC, while other multiple mechanisms also contribute to this progression, such as tumor immunity, cancer stem cells, epithelial–mesenchymal transition and DNA repair disorder. In terms of the therapeutic methods of CRPC, chemotherapy with drugs, such as docetaxel, has been the first-line therapy for CRPC for many years. Besides, newer agents, which target some of the above mechanisms, show additional overall survival benefits for CRPC patients. These therapies include drugs targeting the androgen axis pathway (androgen synthesis, androgen receptor splice variants, coactivators of AR and so on), PI3K-AKT pathway, WNT pathway, DNA repair, rearrangement of ETS gene, novel chemotherapy and immunotherapy, bone metastasis therapy and so on. Understanding these novel findings on the mechanisms of CRPC and the latest potential CRPC therapies will direct us for further exploration of CRPC. Conclusion Through comprehensive consideration, the predominant mechanism of CRPC might be the AR signal axis concomitant with tumor microenvironment, stress, immunity, tumor microenvironment and so on. For CRPC therapy, targeting the AR axis pathway and chemotherapy are the first-line treatments at present. However, with the advancements in CRPC therapy made by the researchers, other novel potential methods will occupy more and more important position in the treatment of CRPC, especially the therapies targeting the tumor microenviroment, tumor immunity and DNA repair and so on.
Collapse
Affiliation(s)
- Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tianbo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
17
|
Lam HM, Corey E. Supraphysiological Testosterone Therapy as Treatment for Castration-Resistant Prostate Cancer. Front Oncol 2018; 8:167. [PMID: 29872642 PMCID: PMC5972313 DOI: 10.3389/fonc.2018.00167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/01/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
18
|
Su W, Sun L, Yang S, Zhao H, Zeng T, Wu W, Wang D. Apolipoprotein C1 promotes prostate cancer cell proliferation in vitro. J Biochem Mol Toxicol 2018; 32:e22158. [PMID: 29719090 PMCID: PMC6099310 DOI: 10.1002/jbt.22158] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/06/2018] [Indexed: 01/07/2023]
Abstract
Here, we aimed to investigate the carcinogenic effects of apolipoprotein C1 (APOC1) in prostate cancer (PCa). APOC1 expression was evaluated in PCa and normal prostate specimens, and lentivirus-mediated RNA interference was used to knockdown APOC1 in DU145 cells. The effects of APOC1 silencing on cell proliferation, cell cycle arrest, and apoptosis were assessed. APOC1 expression was much higher in PCa tissues than in normal tissues. Moreover, APOC1 silencing inhibited cell proliferation and colony formation, arrested cell cycle progression, and enhanced apoptosis in DU145 cells. Additionally, APOC1 silencing decreased survivin, phospho-Rb, and p21 levels and increased cleaved caspase-3 expression. These data supported the procarcinogenic effects of APOC1 in the pathogenesis of PCa and suggested that targeting APOC1 may have applications in the treatment of PCa.
Collapse
Affiliation(s)
- Wei‐peng Su
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| | - Li‐na Sun
- Department of Presbyatrics, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| | - Shun‐liang Yang
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| | - Hu Zhao
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
- Department of Urology, Fuzhou Dongfang HospitalXiamen UniversityXiamenFujian 361005People's Republic of China
| | - Teng‐yue Zeng
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| | - Wei‐zhen Wu
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| | - Dong Wang
- Department of Urology, Fuzhou General HospitalFujian Medical UniversityFuzhouFujian 350025People's Republic of China
| |
Collapse
|
19
|
Yin X, Yu J, Zhou Y, Wang C, Jiao Z, Qian Z, Sun H, Chen B. Identification of CDK2 as a novel target in treatment of prostate cancer. Future Oncol 2018; 14:709-718. [PMID: 29323532 DOI: 10.2217/fon-2017-0561] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM This study aims the potential gene involved in the metastasis of prostate cancer (Pca). METHODS PubMed GEO datasets (GSE6605 and GSE6606) were downloaded. We used multiple bioinformatics methods to screen differentially expressed genes in Pca. Gene network was built by STRING and visualized by Cytoscape. All of the hub genes were analyzed by cBioPortal. Inhibition of CDK2 including siRNA, inhibitor and cas9-induced CDK2 knockout was followed by an invasion assay. Downstream genes of CDK2 were analyzed by western blot. RESULTS Sequencing data were analyzed to screen the genes with expression alterations. The top genes were validated in our samples. 11 hub genes were screened out. Among these genes, STAT3 and CDK2 were significantly associated with recurrence. Further study suggested that inhibition of CDK2 reduced invasion of Pca cell lines. The invasion ability was rescued after reintroduction of CDK2. CONCLUSION These data indicated that CDK2 was a crucial factor in metastasis of Pca and might be a novel therapy target. [Formula: see text].
Collapse
Affiliation(s)
- Xifeng Yin
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jun Yu
- Department of Obstetrics & Gynecology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yang Zhou
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chengyue Wang
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhimin Jiao
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhounan Qian
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hao Sun
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
20
|
Mohammad OS, Nyquist MD, Schweizer MT, Balk SP, Corey E, Plymate S, Nelson PS, Mostaghel EA. Supraphysiologic Testosterone Therapy in the Treatment of Prostate Cancer: Models, Mechanisms and Questions. Cancers (Basel) 2017; 9:E166. [PMID: 29210989 PMCID: PMC5742814 DOI: 10.3390/cancers9120166] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Since Huggins defined the androgen-sensitive nature of prostate cancer (PCa), suppression of systemic testosterone (T) has remained the most effective initial therapy for advanced disease although progression inevitably occurs. From the inception of clinical efforts to suppress androgen receptor (AR) signaling by reducing AR ligands, it was also recognized that administration of T in men with castration-resistant prostate cancer (CRPC) could result in substantial clinical responses. Data from preclinical models have reproducibly shown biphasic responses to T administration, with proliferation at low androgen concentrations and growth inhibition at supraphysiological T concentrations. Many questions regarding the biphasic response of PCa to androgen treatment remain, primarily regarding the mechanisms driving these responses and how best to exploit the biphasic phenomenon clinically. Here we review the preclinical and clinical data on high dose androgen growth repression and discuss cellular pathways and mechanisms likely to be involved in mediating this response. Although meaningful clinical responses have now been observed in men with PCa treated with high dose T, not all men respond, leading to questions regarding which tumor characteristics promote response or resistance, and highlighting the need for studies designed to determine the molecular mechanism(s) driving these responses and identify predictive biomarkers.
Collapse
Affiliation(s)
- Osama S Mohammad
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- Faculty of Medicine, Benha University, Benha 13518, Egypt.
| | | | - Michael T Schweizer
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Stephen P Balk
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA.
| | - Stephen Plymate
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
21
|
Conditionally reprogrammed normal and primary tumor prostate epithelial cells: a novel patient-derived cell model for studies of human prostate cancer. Oncotarget 2017; 8:22741-22758. [PMID: 28009986 PMCID: PMC5410259 DOI: 10.18632/oncotarget.13937] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/22/2016] [Indexed: 01/16/2023] Open
Abstract
Our previous study demonstrated that conditional reprogramming (CR) allows the establishment of patient-derived normal and tumor epithelial cell cultures from a variety of tissue types including breast, lung, colon and prostate. Using CR, we have established matched normal and tumor cultures, GUMC-29 and GUMC-30 respectively, from a patient's prostatectomy specimen. These CR cells proliferate indefinitely in vitro and retain stable karyotypes. Most importantly, only tumor-derived CR cells (GUMC-30) produced tumors in xenografted SCID mice, demonstrating maintenance of the critical tumor phenotype. Characterization of cells with DNA fingerprinting demonstrated identical patterns in normal and tumor CR cells as well as in xenografted tumors. By flow cytometry, both normal and tumor CR cells expressed basal, luminal, and stem cell markers, with the majority of the normal and tumor CR cells expressing prostate basal cell markers, CD44 and Trop2, as well as luminal marker, CD13, suggesting a transit-amplifying phenotype. Consistent with this phenotype, real time RT-PCR analyses demonstrated that CR cells predominantly expressed high levels of basal cell markers (KRT5, KRT14 and p63), and low levels of luminal markers. When the CR tumor cells were injected into SCID mice, the expression of luminal markers (AR, NKX3.1) increased significantly, while basal cell markers dramatically decreased. These data suggest that CR cells maintain high levels of proliferation and low levels of differentiation in the presence of feeder cells and ROCK inhibitor, but undergo differentiation once injected into SCID mice. Genomic analyses, including SNP and INDEL, identified genes mutated in tumor cells, including components of apoptosis, cell attachment, and hypoxia pathways. The use of matched patient-derived cells provides a unique in vitro model for studies of early prostate cancer.
Collapse
|
22
|
Yu P, Duan X, Cheng Y, Liu C, Chen Y, Liu W, Yin B, Wang X, Tao Z. Androgen-independent LNCaP cells are a subline of LNCaP cells with a more aggressive phenotype and androgen suppresses their growth by inducing cell cycle arrest at the G1 phase. Int J Mol Med 2017; 40:1426-1434. [PMID: 28901378 PMCID: PMC5627872 DOI: 10.3892/ijmm.2017.3125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 08/17/2017] [Indexed: 11/06/2022] Open
Abstract
Androgen deprivation therapy (ADT, surgical or chemical castration) is the mainstay treatment for metastatic prostate cancer (PCa); however, patients ineluctably relapse despite extremely low androgen levels. This evolution of PCa indicates its lethal progression. In this study, to mimic the traits of clinical PCa progression in vitro, we investigated the alterations in the cell biological characteristics in androgen-independent LNCaP cells (LNCaP-AI cells) compared with LNCaP cells. We also examined the effects of androgen on LNCaP and LNCaP-AI cell proliferation, androgen receptor (AR) expression and prostate-specific antigen (PSA) secretion. Furthermore, AR was silenced in the LNCaP and LNCaP-AI cells to detect the roles taht AR plays in cell growth, apoptosis and PSA secretion. We found that prolonged androgen ablation increased the LNCaP-AI cell growth rate and cell invasiveness, and induced epithelial-mesenchymal transition in the LNCaP-AI cells. Moreover, despite the fact that the LNCaP and LNCaP-AI cells expressed equal amounts of AR protein, androgen induced a greater secretion of PSA in the LNCaP-AI cells than in the LNCaP cells. The proliferation of the LNCaP-AI cells was not dependent on, but was suppressed by androgen, which led to arrest at the G1 phase. Conversely, androgen significantly increased LNCaP cell proliferation by promoting the G1-S transition. Moreover, the silencing of AR suppressed LNCaP and LNCaP-AI cell growth by inducing cell cycle arrest at the G1 phase rather than promoting apoptosis, and reduced PSA secretion. On the whole, our data suggest that LNCaP-AI cells have a more more aggressive phenotype compared with the LNCaP cells; AR remains a critical factor in the LNCaP-AI cells, and androgen suppresses LNCaP-AI cell growth by blocking the cell cycle at the G1 phase.
Collapse
Affiliation(s)
- Pan Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yue Cheng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Chunhua Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yuhua Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Binbin Yin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
23
|
Schweizer MT, Antonarakis ES, Denmeade SR. Bipolar Androgen Therapy: A Paradoxical Approach for the Treatment of Castration-resistant Prostate Cancer. Eur Urol 2017; 72:323-325. [PMID: 28365160 PMCID: PMC5872811 DOI: 10.1016/j.eururo.2017.03.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/21/2023]
Abstract
Bipolar androgen therapy (BAT) is a paradoxical treatment for castrate-resistant prostate cancer whereby testosterone levels are rapidly alternated between supraphysiologic and near-castrate concentrations. Initial studies demonstrated that BAT is safe and produces clinical responses. A trial comparing enzalutamide against BAT is ongoing.
Collapse
Affiliation(s)
- Michael T Schweizer
- Department of Medicine, Division of Oncology, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Emmanuel S Antonarakis
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Zhou L, Wu F, Jin W, Yan B, Chen X, He Y, Yang W, Du W, Zhang Q, Guo Y, Yuan Q, Dong X, Yu W, Zhang J, Xiao L, Tong P, Shan L, Efferth T. Theabrownin Inhibits Cell Cycle Progression and Tumor Growth of Lung Carcinoma through c-myc-Related Mechanism. Front Pharmacol 2017; 8:75. [PMID: 28289384 PMCID: PMC5326752 DOI: 10.3389/fphar.2017.00075] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022] Open
Abstract
Green tea, the fresh leaves of Camellia sinensis, is not only a health-promoting beverage but also a traditional Chinese medicine used for prevention or treatment of cancer, such as lung cancer. Theabrownin (TB) is the main fraction responsible for the medicinal effects of green tea, but whether it possesses anti-cancer effect is unknown yet. This study aimed to determine the in vitro and in vivo anti-lung cancer effect of TB and explore the underlying molecular mechanism, by using A549 cell line and Lewis lung carcinoma-bearing mice. In cellular experiment, MTT assay was performed to evaluate the inhibitory effect and IC50 values of TB, and flow cytometry was conducted to analyze the cell cycle progression affected by TB. In animal experiment, mice body mass, tumor incidence, tumor size and tumor weight were measured, and histopathological analysis on tumor was performed with Transferase dUTP nick-end labeling staining. Real time PCR and western blot assays were adopted to detect the expression of C-MYC associated genes and proteins for mechanism clarification. TB was found to inhibit A549 cell viability in a dose- and time-dependent manner and block A549 cell cycle at G0/G1 phase. Down-regulation of c-myc, cyclin A, cyclin D, cdk2, cdk4, proliferation of cell nuclear antigen and up-regulation of p21, p27, and phosphate and tension homolog in both gene and protein levels were observed with TB treatment. A c-myc-related mechanism was thereby proposed, since c-myc could transcriptionally regulate all other genes in its downstream region for G1/S transitions of cell cycle and proliferation of cancer cells. This is the first report regarding the anti-NSCLC effect and the underlying mechanism of TB on cell cycle progression and proliferation of A549 cells. The in vivo data verified the in vitro result that TB could significantly inhibit the lung cancer growth in mice and induce apoptosis on tumors in a dose-dependent manner. It provides a promising candidate of natural products for lung cancer therapy and new development of anti-cancer agent.
Collapse
Affiliation(s)
- Li Zhou
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Feifei Wu
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences Inc.Hangzhou, China
| | - Wangdong Jin
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Bo Yan
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Xin Chen
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Yingfei He
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Weiji Yang
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Wenlin Du
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences Inc.Hangzhou, China
| | - Qiang Zhang
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences Inc.Hangzhou, China
| | - Yonghua Guo
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences Inc.Hangzhou, China
| | - Qiang Yuan
- The Second Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhou, China
| | | | - Wenhua Yu
- Hangzhou First People’s HospitalHangzhou, China
| | | | - Luwei Xiao
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Letian Shan
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical UniversityHangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences Inc.Hangzhou, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University of MainzMainz, Germany
| |
Collapse
|
25
|
Palethorpe HM, Drew PA, Smith E. Androgen Signaling in Esophageal Adenocarcinoma Cell Lines In Vitro. Dig Dis Sci 2017; 62:3402-3414. [PMID: 29052817 PMCID: PMC5694516 DOI: 10.1007/s10620-017-4794-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/06/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND We showed previously that nuclear localization of the androgen receptor (AR) and expression of the androgen-responsive gene FK506-binding protein 5 (FKBP5) in esophageal adenocarcinoma (EAC) tissues were associated with decreased patient survival, suggesting a role for androgens in this cancer. AIM To investigate the effect of the AR ligand 5α-dihydrotestosterone (DHT) on AR-expressing EAC cell lines in vitro. METHODS AND RESULTS In tissue resection specimens from EAC patients, FKBP5 expression was positively associated with proliferation as measured by Ki-67 expression. We stably transduced AR into three AR-negative EAC cell lines, OE33, JH-EsoAd1, and OE19, to investigate androgen signaling in vitro. In the AR-expressing cell lines, 10 nM DHT, the concentration typically used to study AR signaling, induced changes in the expression of androgen-responsive genes and inhibited proliferation by inducing cell cycle arrest and senescence. At lower DHT concentrations near the half maximal inhibitory concentration (IC50), the AR-expressing cell lines proliferated and there were changes in the expression of androgen-responsive genes. In direct co-culture with cancer-associated fibroblast-like PShTert myofibroblasts, 10 nM DHT induced changes in the expression of androgen-responsive genes but did not inhibit proliferation. CONCLUSIONS This is the first study to show that EAC cell lines respond to androgen in vitro. Proliferation together with the expression of androgen-responsive genes was dependent on the concentration of DHT, or the presence of a permissive microenvironment, consistent with observations in the tissues. These findings are consistent with a role for androgen signaling in EAC.
Collapse
Affiliation(s)
- Helen M. Palethorpe
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia
| | - Paul A. Drew
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia ,0000 0004 0367 2697grid.1014.4School of Nursing and Midwifery, Flinders University, PO Box 2100, Adelaide, SA 5001 Australia
| | - Eric Smith
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia ,0000 0004 0486 659Xgrid.278859.9Department of Medical Oncology, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville South, SA 5011 Australia
| |
Collapse
|
26
|
Distinct outcomes of CRL-Nedd8 pathway inhibition reveal cancer cell plasticity. Cell Death Dis 2016; 7:e2505. [PMID: 27906189 PMCID: PMC5261022 DOI: 10.1038/cddis.2016.395] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 01/12/2023]
Abstract
Inhibition of protein degradation by blocking Cullin-RING E3 ligases (CRLs) is a new approach in cancer therapy though of unknown risk because CRL inhibition may stabilize both oncoproteins and tumor suppressors. Probing CRLs in prostate cancer cells revealed a remarkable plasticity of cells with TMPRSS2-ERG translocation. CRL suppression by chemical inhibition or knockdown of RING component RBX1 led to reversible G0/G1 cell cycle arrest that prevented cell apoptosis. Conversely, complete blocking of CRLs at a higher inhibitor dose-induced cytotoxicity that was amplified by knockdown of CRL regulator Cand1. We analyzed cell signaling to understand how varying degrees of CRL inhibition translated to distinct cell fates. Both tumor suppressor and oncogenic cell signaling pathways and transcriptional activities were affected, with pro-metastatic Wnt/β-catenin as the most upregulated. Suppression of the NF-κB pathway contributed to anti-apoptotic effect, and androgen receptor (AR) and ERG played decisive, though opposite, roles: AR was involved in protective quiescence, whereas ERG promoted apoptosis. These data define AR–ERG interaction as a key plasticity and survival determinant in prostate cancer and suggest supplementary treatments that may overcome drug resistance mechanisms regulated by AR–ERG interaction.
Collapse
|
27
|
Lin HP, Lin CY, Huo C, Jan YJ, Tseng JC, Jiang SS, Kuo YY, Chen SC, Wang CT, Chan TM, Liou JY, Wang J, Chang WSW, Chang CH, Kung HJ, Chuu CP. AKT3 promotes prostate cancer proliferation cells through regulation of Akt, B-Raf, and TSC1/TSC2. Oncotarget 2016; 6:27097-112. [PMID: 26318033 PMCID: PMC4694976 DOI: 10.18632/oncotarget.4553] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/29/2015] [Indexed: 01/09/2023] Open
Abstract
The qRT-PCR analysis of 139 clinical samples and analysis of 150 on-line database clinical samples indicated that AKT3 mRNA expression level was elevated in primary prostate tumors. Immunohistochemical staining of 65 clinical samples revealed that AKT3 protein expression was higher in prostate tumors of stage I, II, III as compared to nearby normal tissues. Plasmid overexpression of AKT3 promoted cell proliferation of LNCaP, PC-3, DU-145, and CA-HPV-10 human prostate cancer (PCa) cells, while knockdown of AKT3 by siRNA reduced cell proliferation. Overexpression of AKT3 increased the protein expression of total AKT, phospho-AKT S473, phospho-AKT T308, B-Raf, c-Myc, Skp2, cyclin E, GSK3β, phospho-GSK3β S9, phospho-mTOR S2448, and phospho-p70S6K T421/S424, but decreased TSC1 (tuberous sclerosis 1) and TSC2 (tuberous Sclerosis Complex 2) proteins in PC-3 PCa cells. Overexpression of AKT3 also increased protein abundance of phospho-AKT S473, phospho-AKT T308, and B-Raf but decreased expression of TSC1 and TSC2 proteins in LNCaP, DU-145, and CA-HPV-10 PCa cells. Oncomine datasets analysis suggested that AKT3 mRNA level was positively correlated to BRAF. Knockdown of AKT3 in DU-145 cells with siRNA increased the sensitivity of DU-145 cells to B-Raf inhibitor treatment. Knockdown of TSC1 or TSC2 promoted the proliferation of PCa cells. Our observations implied that AKT3 may be a potential therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Hui-Ping Lin
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Ching-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Department of Life Sciences, National Central University, Taiwan
| | - Yee-Jee Jan
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan.,Medical College of Chung Shan Medical University, Taichung City, Taiwan
| | - Jen-Chih Tseng
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Shyh-Chang Chen
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Chih-Ting Wang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Tzu-Min Chan
- Department of Medical Education and Research, China Medical University Beigan Hospital, Yunlin, Taiwan
| | - Jun-Yang Liou
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - John Wang
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Wun-Shaing Wayne Chang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsing-Jien Kung
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung City, Taiwan.,Graduate Program for Aging, China Medical University, Taichung City, Taiwan.,Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung City, Taiwan.,Ph.D. program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| |
Collapse
|
28
|
Anti-Cancer Effect of Lambertianic Acid by Inhibiting the AR in LNCaP Cells. Int J Mol Sci 2016; 17:ijms17071066. [PMID: 27399684 PMCID: PMC4964442 DOI: 10.3390/ijms17071066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 12/13/2022] Open
Abstract
Lambertianic acid (LA) is known to have anti-allergic and antibacterial effects. However, the anticancer activities and mechanism of action of LA have not been investigated. Therefore, the anticancer effects and mechanism of LA are investigated in this study. LA decreased not only AR protein levels, but also cellular and secretory levels of PSA. Furthermore, LA inhibited nuclear translocation of the AR induced by mibolerone. LA suppressed cell proliferation by inducing G1 arrest, downregulating CDK4/6 and cyclin D1 and activating p53 and its downstream molecules, p21 and p27. LA induced apoptosis and the expression of related proteins, including cleaved caspase-9 and -3, c-PARP and BAX, and inhibited BCl-2. The role of AR in LA-induced apoptosis was assessed by using siRNA. Collectively, these findings suggest that LA exerts the anticancer effect by inhibiting AR and is a valuable therapeutic agent in prostate cancer treatment.
Collapse
|
29
|
Androgen receptor inhibits epithelial-mesenchymal transition, migration, and invasion of PC-3 prostate cancer cells. Cancer Lett 2015; 369:103-11. [PMID: 26297988 DOI: 10.1016/j.canlet.2015.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 01/05/2023]
Abstract
Bone metastasis is very common in prostate cancer (PCa) and causes severe pain. PC-3 is an androgen receptor (AR)-negative PCa cell line with high metastatic potential established from PCa bone metastasis. We observed that re-expression of AR, which is located in the cytoplasm in the absence of androgen, suppressed cell motility, migration, and invasion of PC-3 cells as determined by wound healing assay and transwell assay. Micro-Western Array and Western blotting analysis indicated that re-expression of AR increased APC, Akt2, Akt3, PI3K p85, phospho-PI3K p85 Tyr458, PI3K p85, and E-cadherin but decreased GSK-3β, phospho-GSK-3β Ser9, phospho-mTOR Ser2448, Skp2, NF-κB p50, Slug, N-cadherin, β-catenin, vimentin, MMP-9, and Snail. Migration and invasion of PC-3 and PC-3(AR) cells were promoted by EGF or IGF-1 but were suppressed by Casodex. Re-expression of AR reduced the activity of MMP-2 and MMP-9 in PC-3 cells. Our observations suggested that re-expressing AR suppresses migration and invasion of PC-3 cells via regulation of EMT marker proteins and MMP activity.
Collapse
|
30
|
Chen W, Zhong X, Wei Y, Liu Y, Yi Q, Zhang G, He L, Chen F, Liu Y, Luo J. TGF-β Regulates Survivin to Affect Cell Cycle and the Expression of EGFR and MMP9 in Glioblastoma. Mol Neurobiol 2015; 53:1648-1653. [PMID: 25682969 DOI: 10.1007/s12035-015-9121-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/01/2015] [Indexed: 01/05/2023]
Abstract
Transforming growth factor beta (TGF-β) is suggestive of a molecular target for cancer therapy due to its involvement in cell cycle, differentiation, and morphogenesis. Meanwhile, survivin is identified as an apoptosis inhibitor and involved in tumorgenesis. Here, we aimed to investigate the potential associations between TGF-β and survivin in glioblastoma U87 cell line. Survivin small interfering RNA (siRNA), Western blotting, and cell cycle analysis were introduced to detect relevant proteins in TGF-β pathways. In this study, we observed a concentration- and time-dependent increase of survivin expression after treatment with TGF-β1. However, the kinase inhibitors U0126 and LY294002 inhibited the upregulation of survivin in comparison with DMSO. In addition, survivin siRNA effectively abrogated survivin expression in U87 cells, therefore affected cells' entry into the S phase of cell cycle, and then repressed the expression of epidermal growth factor receptor (EGFR) and matrix metalloproteinase 9 (MMP9) in comparison with non-transfection. In conclusion, the present study shows that TGF-β upregulates survivin expression via ERK and PI3K/AKT pathway, leading to glioblastoma cell cycle progression. Thus, the blockade of survivin will allow for the treatment of glioblastoma, partially attributing to the inhibition of EGFR and MMP9 expression.
Collapse
Affiliation(s)
- Wenliang Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Xiao Zhong
- Department of Paediatrics, the Affiliated Xiaolan People's Hospital of Southern Medical University, Guangdong, Zhongshan, 528415, China
| | - Yi Wei
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Yun Liu
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Quan Yi
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Genshui Zhang
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Lishan He
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Fajiang Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Yingping Liu
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China
| | - Jiandong Luo
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, 510182, China. .,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Liao A, Shi R, Jiang Y, Tian S, Li P, Song F, Qu Y, Li J, Yun H, Yang X. SDF-1/CXCR4 Axis Regulates Cell Cycle Progression and Epithelial-Mesenchymal Transition via Up-regulation of Survivin in Glioblastoma. Mol Neurobiol 2014; 53:210-215. [PMID: 25421212 DOI: 10.1007/s12035-014-9006-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022]
Abstract
Stromal cell-derived factor 1 (SDF-1)/CXCR4 ligand-receptor axis is widely recommended as an attractive target for cancer therapy. Meanwhile, epithelial-mesenchymal transition (EMT) process is linked to disease pathophysiology. As one of inhibitors of apoptosis proteins, survivin is implicated in the onset and development of cancer. In the present study, we tried to determine the cause-effect associations between SDF-1/CXCR4 axis and survivin expression in glioblastoma U-251 cell line. Survivin activation and inhibition were induced with exogenous SDF-1 and survivin small interfering RNA (survivin siRNA), respectively. Western blot was used to detect relevant proteins in SDF-1/CXCR4 axis. Western blot analysis revealed that survivin expression in U-251 increased in a dose- and time-dependent manner in response to SDF-1 treatment. However, the interference with MEK/ERK and PI3K/AKT pathway prohibited SDF-1-induced survivin up-regulation. Importantly, survivin knockdown abrogated cell cycle progression and the expression of snail and N-cadherin, compared with non-transfectants. In conclusion, the present study shows that SDF-1 up-regulates survivin via MEK/ERK and PI3K/AKT pathway, leading to cell cycle progression and EMT occurrence dependent on survivin. The blockade of survivin will allow for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Anyan Liao
- Department of Radiation Oncology, Peking University 3rd Hospital, No. 49 Hua Yuan Bei Lu, Beijing, 100191, China.
| | - Ranran Shi
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuliang Jiang
- Department of Radiation Oncology, Peking University 3rd Hospital, No. 49 Hua Yuan Bei Lu, Beijing, 100191, China
| | - Suqing Tian
- Department of Radiation Oncology, Peking University 3rd Hospital, No. 49 Hua Yuan Bei Lu, Beijing, 100191, China
| | - Panpan Li
- Shandong University School of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fuxi Song
- Shandong University School of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yalan Qu
- Shandong University School of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jinna Li
- Department of Radiation Oncology, Peking University 3rd Hospital, No. 49 Hua Yuan Bei Lu, Beijing, 100191, China
| | - Haiqin Yun
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiangshan Yang
- Department of Pathology, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|