1
|
Kurganovs NJ, Engedal N. To eat or not to eat: a critical review on the role of autophagy in prostate carcinogenesis and prostate cancer therapeutics. Front Pharmacol 2024; 15:1419806. [PMID: 38910881 PMCID: PMC11190189 DOI: 10.3389/fphar.2024.1419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Around 1 in 7 men will be diagnosed with prostate cancer during their lifetime. Many strides have been made in the understanding and treatment of this malignancy over the years, however, despite this; treatment resistance and disease progression remain major clinical concerns. Recent evidence indicate that autophagy can affect cancer formation, progression, and therapeutic resistance. Autophagy is an evolutionarily conserved process that can remove unnecessary or dysfunctional components of the cell as a response to metabolic or environmental stress. Due to the emerging importance of autophagy in cancer, targeting autophagy should be considered as a potential option in disease management. In this review, along with exploring the advances made on understanding the role of autophagy in prostate carcinogenesis and therapeutics, we will critically consider the conflicting evidence observed in the literature and suggest how to obtain stronger experimental evidence, as the application of current findings in clinical practice is presently not viable.
Collapse
Affiliation(s)
- Natalie Jayne Kurganovs
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Nikolai Engedal
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Yanar S, Bal Albayrak MG, Kasap M, Akpinar G. From Androgen Dependence to Independence in Prostate Cancer: Unraveling Therapeutic Potential and Proteomic Landscape of Hydroxychloroquine as an Autophagy Inhibitor. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:246-255. [PMID: 38722704 DOI: 10.1089/omi.2024.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Prostate cancer is a major planetary health challenge wherein new ways of thinking drug discovery and therapeutics innovation are much needed. Numerous studies have shown that autophagy inhibition holds a significant role as an adjunctive intervention in prostate cancer. Hydroxychloroquine (HCQ) has gained considerable attention due to its established role as an autophagy inhibitor across diverse cancer types, but its proteomics landscape and systems biology in prostate cancer are currently lacking in the literature. This study reports the proteomic responses to HCQ in prostate cancer cells, namely, androgen-dependent LNCaP and androgen-independent PC3 cells. Differentially expressed proteins and proteome in HCQ-treated cells were determined by label-free quantification with nano-high-performance liquid chromatography and tandem mass spectrometry (nHPLC-MS/MS), and harnessing bioinformatics tools. In PC3 cells, there was a marked shift toward metabolic reprogramming, highlighted by an upregulation of mitochondrial proteins in oxidative phosphorylation and tricarboxylic acid cycle, suggesting an adaptive mechanism to maintain energy production under therapeutic stress. In contrast, LNCaP cells prioritized proteostasis and cell cycle regulation, indicating a more conservative adaptation strategy. To the best of our knowledge, this study is the first to demonstrate the differential responses of prostate cancer cells to autophagy inhibition by HCQ, suggesting that a combination therapy approach, targeting distinct pathways in androgen-independent and androgen-dependent cells, could represent a promising treatment strategy. Moreover, the varied proteomic responses observed between these cell lines underscore the importance of personalized medicine in cancer therapy. Future translational and clinical research on HCQ and prostate cancer are called for.
Collapse
Affiliation(s)
- Sevinc Yanar
- Department of Histology and Embryology, Sakarya University Faculty of Medicine, Sakarya, Turkey
| | | | - Murat Kasap
- Department of Medical Biology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Gurler Akpinar
- Department of Medical Biology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| |
Collapse
|
3
|
Combination of the NRF2 Inhibitor and Autophagy Inhibitor Significantly Inhibited Tumorigenicity of Castration-Resistant Prostate Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4182401. [PMID: 35770119 PMCID: PMC9236814 DOI: 10.1155/2022/4182401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022]
Abstract
Prostate cancer (PCa) is the most frequent cancer in men. Developing new treatment methods for CRPC will be a significant challenge in the clinical treatment of PCa. In conclusion, the results of this study show that NRF2 is downregulated in untreated PCa samples compared to normal PCa samples; however, it was upregulated in mCRPC samples compared to HSPC samples. These results demonstrated that NRF2 may serve as a tumor suppressor in tumorigenesis but promote PCa androgen-independent transferring after ADT treatment. Bioinformatics analysis showed that NRF2 was related to multiple signaling, such as the AGE-RAGE pathway, MAPK pathway, NF-kappa B signaling, PI3K-Akt signaling pathway, and VEGF signaling pathway. Moreover, we revealed that the NRF2 inhibitor significantly inhibited tumorigenicity of CRPC cells in vitro. Of note, combination of the NRF2 inhibitor and autophagy inhibitor had a more significantly suppressive role than either ML385 or CQ, indicating that combination of CQ (autophagy inhibitor) and ML385 (NRF2 inhibitor) is a potential treatment of CRPC. Finally, we conformed that high levels of autophagy regulators LC3B, ULK1, and beclin1 significantly correlated to longer PSA recurrence-free survival time. We think that this study could provide more evidence to confirm that NRF2 is a crucial regulator and targeting NRF2 and autophagy is a potential therapy option for CRPC.
Collapse
|
4
|
Ashrafizadeh M, Paskeh MDA, Mirzaei S, Gholami MH, Zarrabi A, Hashemi F, Hushmandi K, Hashemi M, Nabavi N, Crea F, Ren J, Klionsky DJ, Kumar AP, Wang Y. Targeting autophagy in prostate cancer: preclinical and clinical evidence for therapeutic response. J Exp Clin Cancer Res 2022; 41:105. [PMID: 35317831 PMCID: PMC8939209 DOI: 10.1186/s13046-022-02293-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer is a leading cause of death worldwide and new estimates revealed prostate cancer as the leading cause of death in men in 2021. Therefore, new strategies are pertinent in the treatment of this malignant disease. Macroautophagy/autophagy is a “self-degradation” mechanism capable of facilitating the turnover of long-lived and toxic macromolecules and organelles. Recently, attention has been drawn towards the role of autophagy in cancer and how its modulation provides effective cancer therapy. In the present review, we provide a mechanistic discussion of autophagy in prostate cancer. Autophagy can promote/inhibit proliferation and survival of prostate cancer cells. Besides, metastasis of prostate cancer cells is affected (via induction and inhibition) by autophagy. Autophagy can affect the response of prostate cancer cells to therapy such as chemotherapy and radiotherapy, given the close association between autophagy and apoptosis. Increasing evidence has demonstrated that upstream mediators such as AMPK, non-coding RNAs, KLF5, MTOR and others regulate autophagy in prostate cancer. Anti-tumor compounds, for instance phytochemicals, dually inhibit or induce autophagy in prostate cancer therapy. For improving prostate cancer therapy, nanotherapeutics such as chitosan nanoparticles have been developed. With respect to the context-dependent role of autophagy in prostate cancer, genetic tools such as siRNA and CRISPR-Cas9 can be utilized for targeting autophagic genes. Finally, these findings can be translated into preclinical and clinical studies to improve survival and prognosis of prostate cancer patients. • Prostate cancer is among the leading causes of death in men where targeting autophagy is of importance in treatment; • Autophagy governs proliferation and metastasis capacity of prostate cancer cells; • Autophagy modulation is of interest in improving the therapeutic response of prostate cancer cells; • Molecular pathways, especially involving non-coding RNAs, regulate autophagy in prostate cancer; • Autophagy possesses both diagnostic and prognostic roles in prostate cancer, with promises for clinical application.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey.
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 1417466191, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine University of Tehran, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Francesco Crea
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.,Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Daniel J Klionsky
- Life Sciences Institute & Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Yuzhuo Wang
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Yamamuro T, Nakamura S, Yamano Y, Endo T, Yanagawa K, Tokumura A, Matsumura T, Kobayashi K, Mori H, Enokidani Y, Yoshida G, Imoto H, Kawabata T, Hamasaki M, Kuma A, Kuribayashi S, Takezawa K, Okada Y, Ozawa M, Fukuhara S, Shinohara T, Ikawa M, Yoshimori T. Rubicon prevents autophagic degradation of GATA4 to promote Sertoli cell function. PLoS Genet 2021; 17:e1009688. [PMID: 34351902 PMCID: PMC8341604 DOI: 10.1371/journal.pgen.1009688] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/29/2021] [Indexed: 01/03/2023] Open
Abstract
Autophagy degrades unnecessary proteins or damaged organelles to maintain cellular function. Therefore, autophagy has a preventive role against various diseases including hepatic disorders, neurodegenerative diseases, and cancer. Although autophagy in germ cells or Sertoli cells is known to be required for spermatogenesis and male fertility, it remains poorly understood how autophagy participates in spermatogenesis. We found that systemic knockout mice of Rubicon, a negative regulator of autophagy, exhibited a substantial reduction in testicular weight, spermatogenesis, and male fertility, associated with upregulation of autophagy. Rubicon-null mice also had lower levels of mRNAs of Sertoli cell–related genes in testis. Importantly, Rubicon knockout in Sertoli cells, but not in germ cells, caused a defect in spermatogenesis and germline stem cell maintenance in mice, indicating a critical role of Rubicon in Sertoli cells. In mechanistic terms, genetic loss of Rubicon promoted autophagic degradation of GATA4, a transcription factor that is essential for Sertoli cell function. Furthermore, androgen antagonists caused a significant decrease in the levels of Rubicon and GATA4 in testis, accompanied by elevated autophagy. Collectively, we propose that Rubicon promotes Sertoli cell function by preventing autophagic degradation of GATA4, and that this mechanism could be regulated by androgens. Androgens, known as “male” hormones, stimulate and activate their receptors in various tissues, including testicular cells and skeletal muscle cells, thereby maintaining spermatogenesis and muscle mass. Notably, androgens-dependent maintenance of male reproduction is of particular interest because the incidence of male infertility has increased in recent decades. Previous studies revealed that Androgen receptor knockout in Sertoli cells causes defective spermatogenesis, indicating a crucial role of androgens in Sertoli cells. Another study suggested that fatherhood-dependent downregulation of androgens could decrease male fertility, leading the male to concentrate on parenting existing offspring. However, it remains largely unknown how androgen regulates Sertoli cell function for male reproduction. In the present study, our results suggest that androgens regulate testicular levels of Rubicon, a negative regulator of autophagy, to control autophagic degradation of GATA4 that is required for Sertoli cell function. Because autophagy and androgens participate in various cellular processes, we anticipate that this study will provide a solid evidence for understanding such processes.
Collapse
Affiliation(s)
- Tadashi Yamamuro
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Suita, Osaka, Japan
- * E-mail: (SN); (TY)
| | - Yu Yamano
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Tsutomu Endo
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kyosuke Yanagawa
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ayaka Tokumura
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Hideto Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa, Japan
| | - Yusuke Enokidani
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Gota Yoshida
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hitomi Imoto
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Tsuyoshi Kawabata
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Maho Hamasaki
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Akiko Kuma
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Sohei Kuribayashi
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kentaro Takezawa
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuki Okada
- Laboratory of Pathology and Development, The Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-Ku, Tokyo, Japan
| | - Manabu Ozawa
- Laboratory of Reproductive Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
| | - Shinichiro Fukuhara
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Sakyo-Ku, Kyoto, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Reproductive Systems Biology, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate school of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- * E-mail: (SN); (TY)
| |
Collapse
|
6
|
Hu H, Zhou H, Xu D. A review of the effects and molecular mechanisms of dimethylcurcumin (ASC-J9) on androgen receptor-related diseases. Chem Biol Drug Des 2021; 97:821-835. [PMID: 33277796 DOI: 10.1111/cbdd.13811] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Dimethylcurcumin (ASC-J9) is a curcumin analogue capable of inhibiting prostate cancer cell proliferation. The mechanism is associated with the unique role of ASC-J9 in enhancing androgen receptor (AR) degradation. So far, ASC-J9 has been investigated in typical AR-associated diseases such as prostate cancer, benign prostatic hypertrophy, bladder cancer, renal diseases, liver diseases, cardiovascular diseases, cutaneous wound, spinal and bulbar muscular atrophy, ovarian cancer and melanoma, exhibiting great potentials in disease control. In this review, the effects and molecular mechanisms of ASC-J9 on various AR-associated diseases are summarized. Importantly, the effects of ASC-J9 and AR antagonists enzalutamide/bicalutamide on prostate cancer are compared in detail and crucial differences are highlighted. At last, the pharmacological effects of ASC-J9 are summarized and the future applications of ASC-J9 in AR-associated disease control are discussed.
Collapse
Affiliation(s)
- Hang Hu
- National & Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of Biomass, School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Huan Zhou
- Center for Health Science and Engineering, School of Materials Science and Engineering, Hebei University of Technology, Tianjin, China
| | - Defeng Xu
- National & Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of Biomass, School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| |
Collapse
|
7
|
Tian H, Chou FJ, Tian J, Zhang Y, You B, Huang CP, Yeh S, Niu Y, Chang C. ASC-J9® suppresses prostate cancer cell proliferation and invasion via altering the ATF3-PTK2 signaling. J Exp Clin Cancer Res 2021; 40:3. [PMID: 33390173 PMCID: PMC7780640 DOI: 10.1186/s13046-020-01760-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/03/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Early studies indicated that ASC-J9®, an androgen receptor (AR) degradation enhancer, could suppress the prostate cancer (PCa) progression. Here we found ASC-J9® could also suppress the PCa progression via an AR-independent mechanism, which might involve modulating the tumor suppressor ATF3 expression. METHODS The lentiviral system was used to modify gene expression in C4-2, CWR22Rv1 and PC-3 cells. Western blot and Immunohistochemistry were used to detect protein expression. MTT and Transwell assays were used to test the proliferation and invasion ability. RESULTS ASC-J9® can suppress PCa cell proliferation and invasion in both PCa C4-2 and CWR22Rv1 cells via altering the ATF3 expression. Further mechanistic studies reveal that ASC-J9® can increase the ATF3 expression via decreasing Glutamate-cysteine ligase catalytic (GCLC) subunit expression, which can then lead to decrease the PTK2 expression. Human clinical studies further linked the ATF3 expression to the PCa progression. Preclinical studies using in vivo mouse model also proved ASC-J9® could suppress AR-independent PCa cell invasion, which could be reversed after suppressing ATF3. CONCLUSIONS ASC-J9® can function via altering ATF3/PTK2 signaling to suppress the PCa progression in an AR-independent manner.
Collapse
Affiliation(s)
- Hao Tian
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, 300211, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Fu-Ju Chou
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jing Tian
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, 300211, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yong Zhang
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Bosen You
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Chi-Ping Huang
- Sex Hormone Research Center, Department of Urology, China Medical University, Taichung, 404, Taiwan
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, 300211, China.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Sex Hormone Research Center, Department of Urology, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
8
|
Zhang Q, Xu Y, Zhang Z, Li J, Xia Q, Chen Y. Folliculin deficient renal cancer cells exhibit BRCA1 A complex expression impairment and sensitivity to PARP1 inhibitor olaparib. Gene 2020; 769:145243. [PMID: 33069804 DOI: 10.1016/j.gene.2020.145243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Deficiency of folliculin (FLCN) may lead to renal cell carcinoma (RCC) in patients with Birt-Hogg-Dubé (BHD) disease. In this study, we investigated the cytotoxicity induced by PARP inhibitor olaparib in FLCN deficient RCC cells, and the interaction between FLCN and BRCA1 A complex-regulated DNA repair pathway. METHODS AND MATERIALS FLCN expressing (ACHN and UOK257-F) and FLCN deficient (ACHN-2 and UOK257) cell lines were used in this research. Cell viability was detected by clonogenic assay and MTT assay. Flow cytometry and TUNEL assay were used to detect apoptosis. Autophagy in cells was measured by MDC assay, western blot, and transmission electron microscopy. Co-immunoprecipitation, immunofluorescence and western blot experiments were performed to determine the interaction between FLCN protein and BRCA1 A complex. The in vivo experiments were performed in a xenograft model by inoculating UOK 257 in nude mice. RESULTS RCC cells with FLCN protein deficiency were more sensitive to olaparib treatment than the cells with FLCN expression. Olaparib treatment led to more severe autophagy and apoptosis in FLCN deficient ACHN-2 and UOK257 cells compared to the FLCN expressing ACHN and UOK257-F cells. Decreased BRCA1 A complex expression and disruption of DNA repair ability were detected in FLCN-deficient cells, suggesting that FLCN deficiency impaired BRCA1 A complex expression and sensitized cells to PARP inhibitor olaparib. CONCLUSIONS RCC cells deficient in FLCN are sensitive to olaparib treatment due to the impairment of BRCA1 A complex associated DNA repair ability. The results suggest that PARP inhibitor, such as olaparib, may be a potentially effective therapeutic approach for kidney tumors with deficiency of FLCN protein.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yingkun Xu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zhiyu Zhang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Jianyi Li
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Qinghua Xia
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yougen Chen
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
9
|
Tao H, Shi P, Xuan HY, Ding XS. DNA methyltransferase-1 inactivation of androgen receptor axis triggers homocysteine induced cardiac fibroblast autophagy in diabetic cardiac fibrosis. Arch Biochem Biophys 2020; 692:108521. [PMID: 32800775 DOI: 10.1016/j.abb.2020.108521] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/05/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022]
Abstract
Diabetic cardiac fibrosis is one of the main pathological manifestations of diabetic cardiomyopathy (DCM). Cardiac fibroblast autophagy plays critical roles in diabetic cardiac fibrosis, however, the underlying mechanism of cardiac fibroblast autophagy and diabetic cardiac fibrosis still largely unknown. The aim of the study was to investigate the mechanism of DNMT1 mediated DNA methylation alterations control cardiac fibroblast autophagy in diabetic cardiac fibrosis. We employed streptozotocin (STZ)-induced rats DCM, DCM patient and Hcy induced cardiac fibroblast autophagy. Heart tissue sections were stained with H&E, Sirius Red and Masson's trichrome stain. The expression of DNMT1, AR, Collagen genes mRNA was detected by qRT-PCR. MSP and BSP detected the methylation status of the AR promoter. The expression of DNMT1, AR, Collagen and autophagy-related proteins were detected by Western blotting, Immunofluorescence, Immunohistochemistry. Gain and loss function of AR and DNMT1 in cardiac fibroblast was analyzed. DNMT1 inhibition or knockdown elevated the expression of AR in cardiac fibroblast. Furthermore, we found that AR negatively regulation of Hcy induced cardiac fibroblast autophagy. We demonstrated that DNMT1 enhances cardiac fibroblast autophagy in diabetic cardiac fibrosis through inhibiting AR axis. In conclusion, our results provide new insight into the DNMT1 inactivation of AR axis triggers cardiac fibroblast autophagy in diabetic cardiac fibrosis.
Collapse
Affiliation(s)
- Hui Tao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, PR China; Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Peng Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Hai-Yang Xuan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, PR China.
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, PR China.
| |
Collapse
|
10
|
Qin X, Lu A, Ke M, Zhu W, Ye X, Wang G, Weng G. DJ-1 inhibits autophagy activity of prostate cancer cells by repressing JNK-Bcl2-Beclin1 signaling. Cell Biol Int 2020; 44:937-946. [PMID: 31868268 DOI: 10.1002/cbin.11290] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022]
Abstract
The regulation of DJ-1 on AR signaling plays an important role in the pathogenesis of prostate cancer (PCa). DJ-1 could alter autophagy and regulate Beclin1-involved autophagy response through JNK-dependent pathway. JNK is known to mediate autophagy through Bcl2-Beclin1 complex. Therefore, this study aimed to investigate the significance of autophagy in DJ-1-modulated PCa cells. The current studies showed that DJ-1 overexpression in LNCaP decreased LC3 transformation and autophagosome formation. However, DJ-1 knockdown exerted the opposite effect. Moreover, DJ-1 silencing inhibited survival and promoted death in LNCaP, which was recovered by autophagy inhibition with 3-MA. In addition, DJ-1 overexpression inhibited the phosphorylation of JNK and Bcl2, and the dissociation of Beclin1 and Bcl2; while the effect of silencing DJ-1 was completely opposite. More important, JNK activated by anisimycin inhibited the proliferation and promoted death of DJ-1-overexpressed LNCaP while increasing LC3 transformation and LC3-puncta formation, but these results were reversed by the decrease of Beclin1 (by spautin-1). In contrast, when DJ-1 was silenced, the death of LNCaP, LC3 transformation, and LC3-puncta formation were inhibited by JNK inhibitor SP600125, which promoted cell proliferation. However, Bcl2 inhibition (by ABT737) reversed all the effects of SP600125. Our results suggested that DJ-1 in PCa cells could promote the growth of PCa through autophagy inhibition, and JNK-Bcl2-Beclin1 signaling played an important role in it. The study provided new insights into the role of DJ-1 in the development of PCa.
Collapse
Affiliation(s)
- Xiangcheng Qin
- Department of Urology, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| | - Aimei Lu
- Department of Ultrasonography, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| | - Meilin Ke
- Operating Room, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| | - Weizhi Zhu
- Department of Urology, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| | - Xiaolei Ye
- Department of Cytobiology, Ningbo Institute of Medical Science, Ningbo, 315020, Zhejiang, China
| | - Gang Wang
- Department of Urology, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| | - Guobin Weng
- Department of Urology, Ningbo Urology & Nephrology Hospital, Ningbo, 315192, Zhejiang, China
| |
Collapse
|
11
|
Beretta GL, Zaffaroni N. Androgen Receptor-Directed Molecular Conjugates for Targeting Prostate Cancer. Front Chem 2019; 7:369. [PMID: 31192191 PMCID: PMC6546842 DOI: 10.3389/fchem.2019.00369] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/06/2019] [Indexed: 12/18/2022] Open
Abstract
Due to its central role in the cellular biology of prostate cancer (PC), androgen receptor (AR) still remains an important therapeutic target for fighting this tumor. Several drugs targeting AR have been reported so far, and many new molecules are expected for the future. In spite of their antitumor efficacy, these drugs are not selective for malignant cells and are subjected to AR-mediated activation of drug resistance mechanisms that are responsible for several drawbacks, including systemic toxicity and disease recurrence and metastasis. Among the several strategies considered to overcome these drawbacks, very appealing appears the design of hybrid small-molecule conjugates targeting AR to drive drug action on receptor-positive PC cells. These compounds are designed around on an AR binder, which selectively engages AR with high potency, coupled with a moiety endowed with different pharmacological properties. In this review we focus on two classes of compounds: a) small-molecules and AR-ligand based conjugates that reduce AR expression, which allow down-regulation of AR levels by activating its proteasome-mediated degradation, and b) AR-ligand-based conjugates for targeting small-molecules, in which the AR binder tethers small-molecules, including conventional antitumor drugs (e.g., cisplatin, doxorubicin, histone deacetylase inhibitors, as well as photo-sensitizers) and selectively directs drug action toward receptor-positive PC cells.
Collapse
Affiliation(s)
- Giovanni L Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
12
|
Wang S, Ekoue DN, Raj GV, Kittler R. Targeting the turnover of oncoproteins as a new avenue for therapeutics development in castration-resistant prostate cancer. Cancer Lett 2018; 438:86-96. [PMID: 30217566 PMCID: PMC6186492 DOI: 10.1016/j.canlet.2018.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/23/2018] [Accepted: 09/03/2018] [Indexed: 12/19/2022]
Abstract
The current therapeutic armamentarium for castration-resistant prostate cancer (CRPC) includes second-generation agents such as the Androgen Receptor (AR) inhibitor enzalutamide and the androgen synthesis inhibitor abiraterone acetate, immunotherapies like sipuleucel-T, chemotherapies including docetaxel and cabazitaxel and the radiopharmaceutical radium 223 dichloride. However, relapse of CRPC resistant to these therapeutic modalities occur rapidly. The mechanisms of resistance to these treatments are complex, including specific mutations or alternative splicing of oncogenic proteins. An alternative approach to treating CRPC may be to target the turnover of these molecular drivers of CRPC. In this review, the mechanisms by which protein stability of several oncoproteins such as AR, ERG, GR, CYP17A1 and MYC, will be discussed, as well as how these findings could be translated into novel therapeutic agents.
Collapse
Affiliation(s)
- Shan Wang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Dede N Ekoue
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
13
|
Luo S, Jiang L, Li Q, Sun X, Liu T, Pei F, Zhang T, Liu T, Dong L, Liu X, Jiang L. Acrolein-induced autophagy–dependent apoptosis via activation of the lysosomal–mitochondrial pathway in EAhy926 cells. Toxicol In Vitro 2018; 52:146-153. [DOI: 10.1016/j.tiv.2018.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 05/09/2018] [Accepted: 05/31/2018] [Indexed: 12/18/2022]
|
14
|
Zhao R, Bei X, Yang B, Wang X, Jiang C, Shi F, Wang X, Zhu Y, Jing Y, Han B, Xia S, Jiang Q. Endothelial cells promote metastasis of prostate cancer by enhancing autophagy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:221. [PMID: 30200999 PMCID: PMC6131784 DOI: 10.1186/s13046-018-0884-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Prostate cancer is one of the most common malignancies. Increasing evidence suggested that endothelial cells may contribute to prostate cancer progression and metastasis. Most recently, autophagy has been proposed to plays a significant role in tumorigenesis and metastasis. Also, it is reported that downregulation of androgen receptor (AR) induces autophagy in prostate cancer cells. However, the underlying mechanisms remain unclear. Here, we aim to explore the role and mechanisms of endothelial cell in prostate cancer progression. METHODS The coculture system was established to test the effect of endothelial cells on prostate cancer cells. We performed antibody array and ELISA were used to profile the cytokine expression pattern of endothelial cells in supernatant. Western blot and RT-PCR were used to determine the mechanism by endothelial cells to promote invasion ability of prostate cancer cells. Maraviroc and chloroquine were used to block the CCL5/CCR5 and autophagy pathway respectively. Orthotopic xenograft mouse models and drug treatment study were conducted to determine the role of endothelial cells in promoting metastatic potential in vivo. RESULTS We use CPRC prostate cancer model and demonstrate that endothelial cells secrete large amount of CCL5 and induces autophagy by suppressing AR expression in prostate cancer cell lines. Consequently, elevated autophagy accelerates focal adhesions proteins disassembly and promoted prostate cancer invasion. Inhibition of both CCL5/CCR5 signaling and autophagy significantly reduces metastasis in vivo. CONCLUSIONS Together, our data establish the function for endothelial cells in tumor metastasis and propose new drug target for mCRPC.
Collapse
Affiliation(s)
- Ruizhe Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Xiaoyu Bei
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Boyu Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Chenyi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Xingjie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Yiping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Yifeng Jing
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China.
| | - Qi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, NO, China.
| |
Collapse
|
15
|
Jiang CY, Yang BY, Zhao S, Shao SH, Bei XY, Shi F, Sun Q, Deng Z, Wang XH, Han BM, Zhao FJ, Xia SJ, Ruan Y. Deregulation of ATG9A by impaired AR signaling induces autophagy in prostate stromal fibroblasts and promotes BPH progression. Cell Death Dis 2018; 9:431. [PMID: 29568063 PMCID: PMC5864884 DOI: 10.1038/s41419-018-0415-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 02/04/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
The activation of androgen receptor (AR) signaling plays an essential role in both prostate stromal cells and epithelial cells during the development of benign prostatic hyperplasia (BPH). Here we demonstrated that androgen ablation after 5α-reductase inhibitor (5-ARI) treatment induced autophagy in prostate stromal fibroblasts inhibiting cell apoptosis. In addition, we found that ATG9A expression was increased after androgen ablation, which facilitated autophagic flux development. Knockdown of ATG9A not only inhibited autophagy notably in prostate stromal fibroblasts, but also reduced the volumes of prostate stromal fibroblast and epithelial cell recombinant grafts in nude mice. In conclusion, our findings suggested that ATG9A upregulation after long-term 5-ARI treatment constitutes a possible mechanism of BPH progression. Thus, combined treatment with 5-ARI and autophagy inhibitory agents would reduce the risk of BPH progression.
Collapse
Affiliation(s)
- Chen-Yi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Bo-Yu Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Sheng Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Si-Hui Shao
- Hangzhou Normal University School of Medicine, Hangzhou, 311121, China
| | - Xiao-Yu Bei
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qian Sun
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Zheng Deng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiao-Hai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Fu-Jun Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China.
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
16
|
Fatty acid activated PPARγ promotes tumorigenicity of prostate cancer cells by up regulating VEGF via PPAR responsive elements of the promoter. Oncotarget 2017; 7:9322-39. [PMID: 26814431 PMCID: PMC4891043 DOI: 10.18632/oncotarget.6975] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/17/2016] [Indexed: 11/25/2022] Open
Abstract
In previous work, it is suggested that the excessive amount of fatty acids transported by FABP5 may facilitate the malignant progression of prostate cancer cells through a FABP5-PPARγ-VEGF signal transduction axis to increase angiogenesis. To further functionally characterise the FABP5-PPARγ-VEGF signal transduction pathway, we have, in this work, investigated the molecular mechanisms involved in its tumorigenicity promoting role in prostate cancer. Suppression of PPARγ in highly malignant prostate cancer cells produced a significant reduction (up to 53%) in their proliferation rate, invasiveness (up to 89%) and anchorage-independent growth (up to 94%) in vitro. Knockdown of PPARγ gene in PC3-M cells by siRNA significantly reduced the average size of tumours formed in nude mice by 99% and tumour incidence by 90%, and significantly prolonged the latent period by 3.5 fold. Results in this study combined with some previous results suggested that FABP5 promoted VEGF expression and angiogenesis through PPARγ which was activated by fatty acids transported by FABP5. Further investigations showed that PPARγ up-regulated VEGF expression through acting with the PPAR-responsive elements in the promoter region of VEGF gene in prostate cancer cells. Although androgen can modulate VEGF expression through Sp1/Sp3 binding site on VEGF promoter in androgen-dependent prostate cancer cells, this route, disappeared as the cells gradually lost their androgen dependency; was replaced by the FABP5-PPARγ-VEGF signalling pathway. These results suggested that the FABP5-PPARγ-VEGF signal transduction axis, rather than androgen modulated route, may be a more important novel therapeutic target for angiogenesis-suppression treatment of castration resistant prostate cancer.
Collapse
|
17
|
Chan ML, Liang JW, Hsu LC, Chang WL, Lee SS, Guh JH. Zerumbone, a ginger sesquiterpene, induces apoptosis and autophagy in human hormone-refractory prostate cancers through tubulin binding and crosstalk between endoplasmic reticulum stress and mitochondrial insult. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 388:1223-36. [PMID: 26246051 DOI: 10.1007/s00210-015-1152-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/13/2015] [Indexed: 12/25/2022]
Abstract
Zerumbone, a natural monocyclic sesquiterpene, is the main component of the tropical plant Zingiber zerumbet Smith. Zerumbone induced antiproliferative and apoptotic effects against PC-3 and DU-145, two human hormone-refractory prostate cancer (HRPC) cell lines. Zerumbone inhibited microtubule assembly and induced an increase of MPM-2 expression (specific recognition of mitotic proteins). It also caused an increase of phosphorylation of Bcl-2 and Bcl-xL, two key events in tubulin-binding effect, indicating tubulin-binding capability and mitotic arrest to zerumbone action. Furthermore, zerumbone induced several cellular effects distinct from tubulin-binding properties. First, zerumbone significantly increased, while paclitaxel (as a tubulin-binding control) decreased, Mcl-1 protein expression. Second, paclitaxel but not zerumbone induced Cdk1 activity. Third, zerumbone other than paclitaxel induced Cdc25C downregulation. The data suggest that, in addition to targeting tubulin/microtubule, zerumbone may act on other targets for signaling transduction. Zerumbone induced mitochondrial damage and endoplasmic reticulum (ER) stress as evidenced by the loss of mitochondrial membrane potential and upregulation of GRP-78 and CHOP/GADD153 expression. Zerumbone induced an increase of intracellular Ca(2+) levels, a crosstalk marker between ER stress and mitochondrial insult, associated with the formation of active calpain I fragment. It induced apoptosis through a caspase-dependent way and caused autophagy as evidenced by dramatic LC3-II formation. In summary, the data suggest that zerumbone is a multiple targeting compound that inhibits tubulin assembly and induces a crosstalk between ER stress and mitochondrial insult, leading to apoptosis and autophagy in HRPCs.
Collapse
Affiliation(s)
- Mei-Ling Chan
- School of Pharmacy, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100, Taiwan
| | - Jui-Wei Liang
- School of Pharmacy, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100, Taiwan
| | - Wei-Ling Chang
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Shoei-Sheng Lee
- School of Pharmacy, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100, Taiwan.
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100, Taiwan.
| |
Collapse
|
18
|
Guo J, Huang X, Wang H, Yang H. Celastrol Induces Autophagy by Targeting AR/miR-101 in Prostate Cancer Cells. PLoS One 2015; 10:e0140745. [PMID: 26473737 PMCID: PMC4608724 DOI: 10.1371/journal.pone.0140745] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/30/2015] [Indexed: 01/10/2023] Open
Abstract
Autophagy is an evolutionarily conserved process responsible for the degradation and recycling of cytoplasmic components through autolysosomes. Targeting AR axis is a standard strategy for prostate cancer treatment; however, the role of AR in autophagic processes is still not fully understood. In the present study, we found that AR played a negative role in AR degrader celastrol-induced autophagy. Knockdown of AR in AR-positive prostate cancer cells resulted in enhanced autophagy. Ectopic expression of AR in AR-negative prostate cancer cells, or gain of function of the AR signaling in AR-positive cells, led to suppression of autophagy. Since miR-101 is an inhibitor of autophagy and its expression was decreased along with AR in the process of celastrol-induced autophagy, we hypothesize that AR inhibits autophagy through transactivation of miR-101. AR binding site was defined in the upstream of miR-101 gene by luciferase reporter and ChIP assays. MiR-101 expression correlated with AR status in prostate cancer cell lines. The inhibition of celastrol-induced autophagy by AR was compromised by blocking miR-101; while transfection of miR-101 led to inhibition of celastrol-induced autophagy in spite of AR depletion. Furthermore, mutagenesis of the AR binding site in miR-101 gene led to decreased suppression of autophagy by AR. Finally, autophagy inhibition by miR-101 mimic was found to enhance the cytotoxic effect of celastrol in prostate cancer cells. Our results demonstrate that AR inhibits autophagy via transactivation of miR-101, thus combination of miR-101 mimics with celastrol may represent a promising therapeutic approach for treating prostate cancer.
Collapse
Affiliation(s)
- Jianquan Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Hui Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
- * E-mail:
| |
Collapse
|
19
|
Xie CW, Zhou Y, Liu SL, Fang ZY, Su B, Zhang W. Gabarapl1 mediates androgen-regulated autophagy in prostate cancer. Tumour Biol 2015; 36:8727-33. [PMID: 26050226 DOI: 10.1007/s13277-015-3542-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/06/2015] [Indexed: 11/25/2022] Open
Abstract
Autophagy plays an important role in prostate cancer development. It promotes tumor cell survival and was found to be associated with androgen pathway. In the present study, we found that GABA(A) receptor-associated protein like 1 (Gabarapl1), a ubiquitin-like modifier, participates in the regulation of autophagy. Gabarapl1 is transcriptionally regulated by androgen receptor (AR) and has a repressive role in autophagy. Androgen deprivation downregulates Gabarapl1 in an AR dependent manner, resulting in the increase of autophagy flux. Elevated Gabarapl1 also represses the proliferation of prostate cancer cells. In summary, our study provides evidence to show that Gabarapl1 is a mediator involved in androgen-regulated autophagy process.
Collapse
Affiliation(s)
- Chong-Wei Xie
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - You Zhou
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Sheng-Lin Liu
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Zheng-Yu Fang
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Bing Su
- Xinxiang Key Lab of Translational Cancer Research, The Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, The State University of New York, Buffalo, NY, 14214, USA.
| | - Wei Zhang
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
20
|
Hu Z, Zhang D, Wang D, Sun B, Safoor A, Young CYF, Lou H, Yuan H. Bisbibenzyls, novel proteasome inhibitors, suppress androgen receptor transcriptional activity and expression accompanied by activation of autophagy in prostate cancer LNCaP cells. PHARMACEUTICAL BIOLOGY 2015; 54:364-374. [PMID: 26017567 DOI: 10.3109/13880209.2015.1049278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Bisbibenzyl compounds have gained our interests for their potential antitumor activity in malignant cell-types. OBJECTIVE The objective of this study is to investigate the effect of bisbibenzyl compounds riccardin C (RC), marchantin M (MM), and riccardin D (RD) on androgen receptor (AR) in prostate cancer (PCa) cells. MATERIALS AND METHODS After exposure to 10 μM of the compounds for 24 h, cell cycle and cell survival analyses were performed using FACS and MTT assay to confirm the effect of these bisbibenzyls on PCa LNCaP cells. Changes in the AR expression and function, as the result of exposure to the compounds, were investigated using real-time PCR, ELISA, transient transfection, western blotting (WB), immunoprecipitation, and immunofluorescence staining (IF). Chemical-induced autophagy was examined by WB, IF, and RNAi. RESULTS RC, MM, and RD reduced the viability of LNCaP cells accompanied with arrested cell cycle in the G0/G1 phase and induction of apoptosis. Further investigation revealed that these compounds significantly inhibited AR expression at mRNA and protein levels, leading to the suppression of AR transcriptional activity. Moreover, inhibition of proteasome activity by bisbibenzyls, which in turn caused the induction of autophagy, as noted by induction of LC3B expression, conversion, and accumulation of punctate dots in treated cells. Co-localization of AR/LC3B and AR/Ub suggested that autophagy contributed to the degradation of polyubiquitinated-AR when proteasome activity was suppressed by the bisbibenzyls. DISCUSSION AND CONCLUSION Suppression of proteasome activity and induction of autophagy were involved in bisbibenzyl-mediated modulation of AR activities and apoptosis, suggesting their potential in treating PCa.
Collapse
Affiliation(s)
- Zhongyi Hu
- a Department of Biochemistry and Molecular Biology , Shandong University School of Medicine , Jinan , China
| | - Denglu Zhang
- a Department of Biochemistry and Molecular Biology , Shandong University School of Medicine , Jinan , China
| | - Dawei Wang
- a Department of Biochemistry and Molecular Biology , Shandong University School of Medicine , Jinan , China
| | - Bin Sun
- b Department of Natural Product Chemistry , Shandong University School of Pharmaceutical Sciences , Jinan , China , and
| | - Ayesha Safoor
- a Department of Biochemistry and Molecular Biology , Shandong University School of Medicine , Jinan , China
| | - Charles Y F Young
- c Department of Urology , Mayo Clinic College of Medicine, Mayo Clinic , Rochester , MN , USA
| | - Hongxiang Lou
- b Department of Natural Product Chemistry , Shandong University School of Pharmaceutical Sciences , Jinan , China , and
| | - Huiqing Yuan
- a Department of Biochemistry and Molecular Biology , Shandong University School of Medicine , Jinan , China
| |
Collapse
|
21
|
Chandra V, Bhagyaraj E, Parkesh R, Gupta P. Transcription factors and cognate signalling cascades in the regulation of autophagy. Biol Rev Camb Philos Soc 2015; 91:429-51. [PMID: 25651938 DOI: 10.1111/brv.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 01/04/2015] [Accepted: 01/11/2015] [Indexed: 12/11/2022]
Abstract
Autophagy is a process that maintains the equilibrium between biosynthesis and the recycling of cellular constituents; it is critical for avoiding the pathophysiology that results from imbalance in cellular homeostasis. Recent reports indicate the need for the design of high-throughput screening assays to identify targets and small molecules for autophagy modulation. For such screening, however, a better understanding of the regulation of autophagy is essential. In addition to regulation by various signalling cascades, regulation of gene expression by transcription factors is also critical. This review focuses on the various transcription factors as well as the corresponding signalling molecules that act together to translate the stimuli to effector molecules that up- or downregulate autophagy. This review rationalizes the importance of these transcription factors functioning in tandem with cognate signalling molecules and their interfaces as possible therapeutic targets for more specific pharmacological interventions.
Collapse
Affiliation(s)
- Vemika Chandra
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Ella Bhagyaraj
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Raman Parkesh
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Pawan Gupta
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| |
Collapse
|
22
|
Kim JY, Jung WH, Koo JS. Expression of autophagy-related proteins according to androgen receptor and HER-2 status in estrogen receptor-negative breast cancer. PLoS One 2014; 9:e105666. [PMID: 25140630 PMCID: PMC4139390 DOI: 10.1371/journal.pone.0105666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 07/27/2014] [Indexed: 01/07/2023] Open
Abstract
PURPOSE The purpose of this study was to investigate the expression of autophagy-related proteins in relation to androgen receptor (AR) status in estrogen receptor (ER)-negative breast cancers. METHODS We extracted 334 ER-negative breast cancer samples to construct tissue microarrays (TMAs), which were immunohistochemically stained for autophagy-related proteins (beclin-1, LC3A, LC3B, p62) and for AR and HER-2. RESULTS There were 127 AR-positive cases and 207 AR-negative cases, and 140 HER-2-positive cases and 194 HER-2 negative cases. The AR-negative group was associated with tumoral LC3A expression (P<0.001), while the AR-positive group was associated with tumoral BNIP3 expression (P<0.001). Tumoral LC3A was most highly expressed in the AR-negative and HER-2 negative group, while stromal LC3A showed the highest expression in the AR-negative and HER-2-positive group. Tumoral BNIP3 and stromal BNIP3 were highest in the AR-positive and HER-2-negative group. In the AR-positive and HER-2-negative group, stromal p62 positivity was an independent factor that was statistically significant in its association with shorter disease-free survival (DFS) (Hazard ratio: 10.21, 95% CI: 1.130-92.31, P = 0.039). Shorter DFS was associated with tumoral LC3A positivity (Hazard ratio: 10.28, 95% CI: 2.068-51.19, P = 0.004) in the AR-negative and HER-2-positive group. CONCLUSION In ER-negative breast cancers, AR status was associated with expression of different types of autophagy-related proteins. Tumoral LC3A was most highly expressed in AR-negative breast cancers, while tumor BNIP3 was highest in AR-positive breast cancers.
Collapse
Affiliation(s)
- Ji-Ye Kim
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Hee Jung
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ja Seung Koo
- Department of Pathology, Severance Hospital, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
23
|
Abstract
Autophagy, or 'self-eating', is an adaptive process that enables cells to cope with metabolic, toxic, and even infectious stressors. Although the adaptive capability of autophagy is generally considered beneficial, autophagy can also enhance nutrient utilization and improve growth characteristics of cancer cells. Moreover, autophagy can promote greater cellular robustness in the context of therapeutic intervention. In advanced prostate cancer, preclinical data provide evidence that autophagy facilitates both disease progression and therapeutic resistance. Notably, androgen deprivation therapy, taxane-based chemotherapy, targeted kinase inhibition, and nutrient restriction all induce significant cellular distress and, subsequently, autophagy. Understanding the context-dependent role of autophagy in cancer development and treatment resistance has the potential to improve current treatment of advanced prostate cancer. Indeed, preclinical studies have shown that the pharmacological inhibition of autophagy (with agents including chloroquine, hydroxychloroquine, metformin, and desmethylclomipramine) can enhance the cell-killing effect of cancer therapeutics, and a number of these agents are currently under investigation in clinical trials. However, many of these autophagy modulators are relatively nonspecific, and cytotoxicity in noncancerous tissues is still a concern. Moving forward, refinement of autophagy modulation is needed.
Collapse
|
24
|
Verderio P, Pandolfi L, Mazzucchelli S, Marinozzi MR, Vanna R, Gramatica F, Corsi F, Colombo M, Morasso C, Prosperi D. Antiproliferative Effect of ASC-J9 Delivered by PLGA Nanoparticles against Estrogen-Dependent Breast Cancer Cells. Mol Pharm 2014; 11:2864-75. [DOI: 10.1021/mp500222k] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Paolo Verderio
- Dipartimento
di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
- Nerviano Medical Sciences s.r.l., Viale Pasteur 10, 20014 Nerviano, Italy
| | - Laura Pandolfi
- Dipartimento
di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
- Dipartimento
di Scienze Biomediche e Cliniche “Luigi Sacco”, Università di Milano, Ospedale L. Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Serena Mazzucchelli
- Dipartimento
di Scienze Biomediche e Cliniche “Luigi Sacco”, Università di Milano, Ospedale L. Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Maria Rosaria Marinozzi
- Dipartimento
di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
| | - Renzo Vanna
- LABION
- Laboratory of Nanomedicine and Clinical Biophotonics, Fondazione Don Carlo Gnocchi ONLUS, Piazzale R. Morandi 6, 20121 Milano, Italy
| | - Furio Gramatica
- LABION
- Laboratory of Nanomedicine and Clinical Biophotonics, Fondazione Don Carlo Gnocchi ONLUS, Piazzale R. Morandi 6, 20121 Milano, Italy
| | - Fabio Corsi
- Dipartimento
di Scienze Biomediche e Cliniche “Luigi Sacco”, Università di Milano, Ospedale L. Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
| | - Miriam Colombo
- Dipartimento
di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
| | - Carlo Morasso
- LABION
- Laboratory of Nanomedicine and Clinical Biophotonics, Fondazione Don Carlo Gnocchi ONLUS, Piazzale R. Morandi 6, 20121 Milano, Italy
| | - Davide Prosperi
- Dipartimento
di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Piazza
della Scienza 2, 20126 Milano, Italy
- Dipartimento
di Scienze Biomediche e Cliniche “Luigi Sacco”, Università di Milano, Ospedale L. Sacco, Via G.B. Grassi 74, 20157 Milano, Italy
- LABION
- Laboratory of Nanomedicine and Clinical Biophotonics, Fondazione Don Carlo Gnocchi ONLUS, Piazzale R. Morandi 6, 20121 Milano, Italy
| |
Collapse
|
25
|
Zhang Q, Si S, Schoen S, Jin XB, Chen J, Wu G. Folliculin Deficient Renal Cancer Cells Show Higher Radiosensitivity through Autophagic Cell Death. J Urol 2014; 191:1880-8. [DOI: 10.1016/j.juro.2014.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Qi Zhang
- Minimally Invasive Urology Center, Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China
- Department of Urology, University of Rochester Medical Center, Rochester, New York
| | - Shuhui Si
- Department of Urology, University of Rochester Medical Center, Rochester, New York
| | - Sue Schoen
- Department of Urology, University of Rochester Medical Center, Rochester, New York
| | - Xun-Bo Jin
- Minimally Invasive Urology Center, Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| | - Jindong Chen
- Department of Urology, University of Rochester Medical Center, Rochester, New York
| | - Guan Wu
- Department of Urology, University of Rochester Medical Center, Rochester, New York
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
26
|
Atala A. Re: TALEN-Engineered AR Gene Rearrangements Reveal Endocrine Uncoupling of Androgen Receptor in Prostate Cancer. J Urol 2014; 191:1927. [DOI: 10.1016/j.juro.2014.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2014] [Indexed: 10/25/2022]
|
27
|
Nguyen HG, Yang JC, Kung HJ, Shi XB, Tilki D, Lara PN, DeVere White RW, Gao AC, Evans CP. Targeting autophagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model. Oncogene 2014; 33:4521-30. [PMID: 24662833 PMCID: PMC4155805 DOI: 10.1038/onc.2014.25] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/05/2013] [Accepted: 12/13/2013] [Indexed: 01/07/2023]
Abstract
Macro-autophagy is associated with drug resistance in various cancers and can function as an adaptive response to maintain cell survival under metabolic stresses, including androgen deprivation. Androgen deprivation or treatment with androgen receptor (AR) signaling inhibitor (ARSI), Enzalutamide (MDV-3100, ENZA) or bicalutamide induced autophagy in androgen-dependent and in castration-resistant CaP (castration-resistant prostate cancer (CRPC)) cell lines. The autophagic cascade triggered by AR blockage, correlated with the increased light chain 3-II/I ratio and ATG-5 expression. Autophagy was observed in a subpopulation of C4-2B cells that developed insensitivity to ENZA after sustained exposure in culture. Using flow cytometry and clonogenic assays, we showed that inhibiting autophagy with clomipramine (CMI), chloroquine or metformin increased apoptosis and significantly impaired cell viability. This autophagic process was mediated by AMP-dependent protein kinase (AMPK) activation and the suppression of mammalian target of rapamycin (mTOR) through Raptor phosphorylation (Serine 792). Furthermore, small interfering RNA targeting AMPK significantly inhibited autophagy and promoted cell death in CaP cells acutely or chronically exposed to ENZA or androgen deprivation, suggesting that autophagy is an important survival mechanism in CRPC. Lastly, in vivo studies with mice orthotopically implanted with ENZA-resistant cells demonstrated that the combination of ENZA and autophagy modulators, CMI or metformin significantly reduced tumor growth when compared with control groups (P<0.005). In conclusion, autophagy is as an important mechanism of resistance to ARSI in CRPC. Antiandrogen-induced autophagy is mediated through the activation of AMPK pathway and the suppression of mTOR pathway. Blocking autophagy pharmacologically or genetically significantly impairs prostate cancer cell survival in vitro and in vivo, implying the therapeutics potential of autophagy inhibitors in the antiandrogen-resistance setting.
Collapse
Affiliation(s)
- H G Nguyen
- Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA
| | - J C Yang
- Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA
| | - H-J Kung
- 1] Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, USA [2] UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - X-B Shi
- Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA
| | - D Tilki
- Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA
| | - P N Lara
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - R W DeVere White
- 1] Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA [2] UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - A C Gao
- 1] Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA [2] UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - C P Evans
- 1] Department of Urology, UC Davis School of Medicine, Sacramento, CA, USA [2] UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
28
|
Felgueiras J, Silva JV, Fardilha M. Prostate cancer: the need for biomarkers and new therapeutic targets. J Zhejiang Univ Sci B 2014; 15:16-42. [PMID: 24390742 PMCID: PMC3891116 DOI: 10.1631/jzus.b1300106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/08/2013] [Indexed: 12/16/2022]
Abstract
Prostate cancer (PCa) incidence and mortality have decreased in recent years. Nonetheless, it remains one of the most prevalent cancers in men, being a disquieting cause of men's death worldwide. Changes in many cell signaling pathways have a predominant role in the onset, development, and progression of the disease. These include prominent pathways involved in the growth, apoptosis, and angiogenesis of the normal prostate gland, such as androgen and estrogen signaling, and other growth factor signaling pathways. Understanding the foundations of PCa is leading to the discovery of key molecules that could be used to improve patient management. The ideal scenario would be to have a panel of molecules, preferably detectable in body fluids, that are specific and sensitive biomarkers for PCa. In the early stages, androgen deprivation is the gold standard therapy. However, as the cancer progresses, it eventually becomes independent of androgens, and hormonal therapy fails. For this reason, androgen-independent PCa is still a major therapeutic challenge. By disrupting specific protein interactions or manipulating the expression of some key molecules, it might be possible to regulate tumor growth and metastasis formation, avoiding the systemic side effects of current therapies. Clinical trials are already underway to assess the efficacy of molecules specially designed to target key proteins or protein interactions. In this review, we address that recent progress made towards understanding PCa development and the molecular pathways underlying this pathology. We also discuss relevant molecular markers for the management of PCa and new therapeutic challenges.
Collapse
|
29
|
Zhang Q, Si S, Schoen S, Chen J, Jin XB, Wu G. Suppression of autophagy enhances preferential toxicity of paclitaxel to folliculin-deficient renal cancer cells. J Exp Clin Cancer Res 2013; 32:99. [PMID: 24305604 PMCID: PMC3879005 DOI: 10.1186/1756-9966-32-99] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/21/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Paclitaxel, a widely used chemotherapeutic drug, can induce apoptosis in variety of cancer cells. A previous study has shown preferential toxicity of paclitaxel to FLCN-deficient kidney cancer cell line, UOK257. In this report, we investigate the cellular and molecular mechanism of paclitaxel-induced autophagy and apoptosis in renal cancer cells with and without FLCN expression. METHODS Two pairs of cell lines were used: FLCN siRNA-silenced ACHN cell line (ACHN-5968) and scrambled ACHN cell line (ACHN-sc); FLCN-null UOK257 cell line and UOK257-2 cell line restored with ectopic expression of FLCN. Autophagy was examined by western blot, GFP-LC3, transmission electron microscopy, and MDC assay. Cell viability and apoptosis were detected using MTT assay, DAPI stain and TUNEL assay. After inhibition of autophagy with 3-Methyladenine (3-MA) or Beclin 1 siRNA, cell viability and apoptosis were measured by MTT assay and TUNEL assay. RESULTS After paclitaxel treatment, a dose-dependent decrease in cell viability and increase in apoptosis were observed in FLCN-deficient UOK257 and ACHN-5968 cells compared to their FLCN-expressing counterparts, suggesting that renal cancer cells without FLCN were more sensitive to paclitaxel. Enhanced autophagy was found to be associated with paclitaxel treatment in FLCN-deficient RCC cells. The MAPK pathway was also identified as a key pathway for the activation of autophagy in these kidney cancer cells. Inhibition of phosphorylated ERK with ERK inhibitor U0126 showed a significant decrease in autophagy. Furthermore, after inhibition of autophagy with 3-Methyladenine (3-MA) or Beclin 1 siRNA, apoptosis induced by paclitaxel was significantly increased in FLCN-deficient UOK257 and ACHN-5968 cells. CONCLUSIONS Preferential toxicity of paclitaxel to FLCN-deficient kidney cancer cells is associated with enhanced autophagy. Suppression of autophagy further enhances paclitaxel-induced apoptosis in FLCN-deficient renal cancer cells. Our results suggest that paclitaxel combined with an autophagy inhibitor might be a potentially more effective chemotherapeutic approach for FLCN-deficient renal cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Minimally Invasive Urology Center, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 656, 14642 Rochester, NY, USA
| | - Shuhui Si
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 656, 14642 Rochester, NY, USA
| | - Sue Schoen
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 656, 14642 Rochester, NY, USA
| | - Jindong Chen
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 656, 14642 Rochester, NY, USA
| | - Xun-Bo Jin
- Minimally Invasive Urology Center, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Guan Wu
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 656, 14642 Rochester, NY, USA
- Pathology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
30
|
Androgen receptor (AR) positive vs negative roles in prostate cancer cell deaths including apoptosis, anoikis, entosis, necrosis and autophagic cell death. Cancer Treat Rev 2013; 40:31-40. [PMID: 23993415 DOI: 10.1016/j.ctrv.2013.07.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/20/2013] [Accepted: 07/29/2013] [Indexed: 12/21/2022]
Abstract
Androgen/androgen receptor (AR) signaling plays pivotal roles in the prostate development and homeostasis as well as in the progression of prostate cancer (PCa). Androgen deprivation therapy (ADT) with anti-androgens remains as the main treatment for later stage PCa, and it has been shown to effectively suppress PCa growth during the first 12-24 months. However, ADT eventually fails and tumors may re-grow and progress into the castration resistant stage. Recent reports revealed that AR might play complicated and even opposite roles in PCa progression that might depend on cell types and tumor stages. Importantly, AR may influence PCa progression via differential modulation of various cell deaths including apoptosis, anoikis, entosis, necrosis, and autophagic cell deaths. Targeting AR may induce PCa cell apoptosis, autophagic cell deaths and programmed necrosis, yet targeting AR may suppress cell deaths via anoikis and entosis that may potentially lead to increased metastasis. These differential functions of AR in various types of PCa cell death might challenge the current ADT with anti-androgens treatment. Further detailed dissection of molecular mechanisms by which AR modulates different PCa cell deaths will help us to develop a better therapy to battle PCa.
Collapse
|
31
|
Li M, Yang X, Wang H, Xu E, Xi Z. Inhibition of androgen induces autophagy in benign prostate epithelial cells. Int J Urol 2013; 21:195-9. [PMID: 23819759 DOI: 10.1111/iju.12210] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 05/24/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE 5-α Reductase inhibitor can reduce the volume of benign prostatic hyperplasia by lowering benign prostatic hyperplasia level and consequently inducing epithelial cells apoptosis. The present study investigated whether autophagy and apoptosis of benign prostatic hyperplasia epithelial cells are influenced by low benign prostatic hyperplasia levels. METHODS PWR-1E prostate epithelial cells transfected with GFP-LC3 plasmid were subjected to androgen deprivation conditions. Then the autophagic puncta were evaluated by fluorescence microscopy, and the cellular apoptosis rate was detected by 4, 6-diamidino-2-phenylindole staining after blocking of autophagic process by 3-methyladenine. Furthermore, autophagy status was also determined in hyperplasia prostate tissues from 5-α reductase inhibitor-treated patients by immunohistochemistry. RESULTS In the androgen deprivation medium, autophagic punta increased markedly in PWR-1E cells, and blockage of autophagy by 3-methyladenine significantly promoted PWR-1E cells' apoptosis rate. In vivo, the expression of LC3 protein (an important autophagic marker) in hyperplasia prostate tissue significantly increased after 5-α reductase inhibitor treatment. Meanwhile, the prostate-specific antigen, as an inner control, decreased. CONCLUSION 5-α Reductase inhibitor treatment increases autophagy and possibly decreases the apoptosis of prostate epithelial cells.
Collapse
Affiliation(s)
- Mengqiang Li
- Department of Urology, Union Hospital of Fujian Medical University, Fuzhou, China
| | | | | | | | | |
Collapse
|