1
|
Stefanova ME, Ing-Simmons E, Stefanov S, Flyamer I, Dorado Garcia H, Schöpflin R, Henssen AG, Vaquerizas JM, Mundlos S. Doxorubicin Changes the Spatial Organization of the Genome around Active Promoters. Cells 2023; 12:2001. [PMID: 37566080 PMCID: PMC10417312 DOI: 10.3390/cells12152001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
In this study, we delve into the impact of genotoxic anticancer drug treatment on the chromatin structure of human cells, with a particular focus on the effects of doxorubicin. Using Hi-C, ChIP-seq, and RNA-seq, we explore the changes in chromatin architecture brought about by doxorubicin and ICRF193. Our results indicate that physiologically relevant doses of doxorubicin lead to a local reduction in Hi-C interactions in certain genomic regions that contain active promoters, with changes in chromatin architecture occurring independently of Top2 inhibition, cell cycle arrest, and differential gene expression. Inside the regions with decreased interactions, we detected redistribution of RAD21 around the peaks of H3K27 acetylation. Our study also revealed a common structural pattern in the regions with altered architecture, characterized by two large domains separated from each other. Additionally, doxorubicin was found to increase CTCF binding in H3K27 acetylated regions. Furthermore, we discovered that Top2-dependent chemotherapy causes changes in the distance decay of Hi-C contacts, which are driven by direct and indirect inhibitors. Our proposed model suggests that doxorubicin-induced DSBs cause cohesin redistribution, which leads to increased insulation on actively transcribed TAD boundaries. Our findings underscore the significant impact of genotoxic anticancer treatment on the chromatin structure of the human genome.
Collapse
Affiliation(s)
- Maria E. Stefanova
- Development and Disease Research Group, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany (S.M.)
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Elizabeth Ing-Simmons
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; (E.I.-S.); (J.M.V.)
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Stefan Stefanov
- Berlin Institute for Molecular and Systems Biology, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany;
- Department of Biology, Chemistry, and Pharmacology, Institute of Biochemistry, Freie Universität Berlin, 14163 Berlin, Germany
| | - Ilya Flyamer
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland;
| | - Heathcliff Dorado Garcia
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, 13125 Berlin, Germany; (H.D.G.); (A.G.H.)
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Robert Schöpflin
- Development and Disease Research Group, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany (S.M.)
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Anton G. Henssen
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, 13125 Berlin, Germany; (H.D.G.); (A.G.H.)
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Juan M. Vaquerizas
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; (E.I.-S.); (J.M.V.)
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Stefan Mundlos
- Development and Disease Research Group, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany (S.M.)
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
2
|
Jeon KH, Park S, Shin JH, Jung AR, Hwang SY, Seo SH, Jo H, Na Y, Kwon Y. Synthesis and evaluation of 7-(3-aminopropyloxy)-substituted flavone analogue as a topoisomerase IIα catalytic inhibitor and its sensitizing effect to enzalutamide in castration-resistant prostate cancer cells. Eur J Med Chem 2023; 246:114999. [PMID: 36493620 DOI: 10.1016/j.ejmech.2022.114999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Prostate cancer patients primarily receive androgen receptor (AR)-targeted drugs as a primary treatment option because prostate cancer is associated with highly activated AR signaling. AR amplification made prostate cancer cells viable under treatment of AR-targeted therapy, leading to castration resistance. AR amplification was more common in enzalutamide-resistant patients. As a strategy to overcome castration resistance and to improve the efficacy of enzalutamide, second-generation nonsteroidal antiandrogen drugs for castration-resistant prostate cancer (CRPC) including topoisomerase II (topo II) poisons such as etoposide and mitoxantrone, have been administered in combination with enzalutamide. In the present study, it was confirmed that amplification of topo IIα, but not I and IIβ, was directly and proportionally associated with poor clinical outcome of Prostate cancer. Among a novel series of newly designed and synthesized 7-(3-aminopropyloxy)-substituted flavone analogues, compound 6, the most potent derivative, was further characterized and identified as a topo IIα catalytic inhibitor that intercalates into DNA and binds to the DNA minor groove with better efficacy and less genotoxicity than etoposide, a topo II poison. Compound 6 showed remarkable efficacy in inhibiting AR-negative CRPC cell growth and sensitizing activity to enzalutamide in AR-positive CRPC cells, thus confirming the potential of topo IIα catalytic inhibitor to overcome resistance to androgen deprivation therapy.
Collapse
Affiliation(s)
- Kyung-Hwa Jeon
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Jae-Ho Shin
- College of Pharmacy, CHA University, 120 Haeryong-ro, Pochon-shi, Gyeongghi-do, 11160, Republic of Korea
| | - Ah-Reum Jung
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Soo-Yeon Hwang
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Seung Hee Seo
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Hyunji Jo
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Younghwa Na
- College of Pharmacy, CHA University, 120 Haeryong-ro, Pochon-shi, Gyeongghi-do, 11160, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
3
|
Matias-Barrios VM, Dong X. The Implication of Topoisomerase II Inhibitors in Synthetic Lethality for Cancer Therapy. Pharmaceuticals (Basel) 2023; 16:94. [PMID: 36678591 PMCID: PMC9866718 DOI: 10.3390/ph16010094] [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: 11/10/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
DNA topoisomerase II (Top2) is essential for all eukaryotic cells in the regulation of DNA topology through the generation of temporary double-strand breaks. Cancer cells acquire enhanced Top2 functions to cope with the stress generated by transcription and DNA replication during rapid cell division since cancer driver genes such as Myc and EZH2 hijack Top2 in order to realize their oncogenic transcriptomes for cell growth and tumor progression. Inhibitors of Top2 are therefore designed to target Top2 to trap it on DNA, subsequently causing protein-linked DNA breaks, a halt to the cell cycle, and ultimately cell death. Despite the effectiveness of these inhibitors, cancer cells can develop resistance to them, thereby limiting their therapeutic utility. To maximize the therapeutic potential of Top2 inhibitors, combination therapies to co-target Top2 with DNA damage repair (DDR) machinery and oncogenic pathways have been proposed to induce synthetic lethality for more thorough tumor suppression. In this review, we will discuss the mode of action of Top2 inhibitors and their potential applications in cancer treatments.
Collapse
Affiliation(s)
- Victor M. Matias-Barrios
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| | - Xuesen Dong
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
4
|
Hongo H, Kosaka T, Suzuki Y, Mikami S, Fukada J, Oya M. Topoisomerase II alpha inhibition can overcome taxane-resistant prostate cancer through DNA repair pathways. Sci Rep 2021; 11:22284. [PMID: 34782700 PMCID: PMC8593019 DOI: 10.1038/s41598-021-01697-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/02/2021] [Indexed: 11/19/2022] Open
Abstract
Cabazitaxel (CBZ) is approved for the treatment of docetaxel-resistant castration-resistant prostate cancer (CRPC). However, its efficacy against CRPC is limited, and there are no effective treatments for CBZ-resistant CRPC. This study explored the optimal treatment for CRPC in the post-cabazitaxel setting. PC3 (CBZ-sensitive) and PC3CR cells (CBZ-resistant) were used in this study. We performed in silico drug screening for candidate drugs that could reprogram the gene expression signature of PC3CR cells. The in vivo effect of the drug combination was tested in xenograft mice models. We identified etoposide (VP16) as a promising treatment candidate for CBZ-resistant CRPC. The WST assay revealed that VP16 had a significant antitumor effect on PC3CR cells. PC3CR cells exhibited significantly higher topoisomerase II alpha (TOP2A) expression than PC3 cells. Higher TOP2A expression was a poor prognostic factor in The Cancer Genome Atlas prostate cancer cohort. In the Fred Hutchinson Cancer Research Center dataset, docetaxel-exposed tissues and metastatic tumors had higher TOP2A expression. In addition, VP16 significantly inhibited the growth of tumors generated from both cell lines. Based on these findings, VP16-based chemotherapy may be an optimal treatment for CPRC in the post-CBZ setting.
Collapse
Affiliation(s)
- Hiroshi Hongo
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Yoko Suzuki
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shuji Mikami
- Department of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Junichi Fukada
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
5
|
Radaeva M, Ton AT, Hsing M, Ban F, Cherkasov A. Drugging the 'undruggable'. Therapeutic targeting of protein-DNA interactions with the use of computer-aided drug discovery methods. Drug Discov Today 2021; 26:2660-2679. [PMID: 34332092 DOI: 10.1016/j.drudis.2021.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 07/17/2021] [Indexed: 02/09/2023]
Abstract
Transcription factors (TFs) act as major oncodrivers in many cancers and are frequently regarded as high-value therapeutic targets. The functionality of TFs relies on direct protein-DNA interactions, which are notoriously difficult to target with small molecules. However, this prior view of the 'undruggability' of protein-DNA interfaces has shifted substantially in recent years, in part because of significant advances in computer-aided drug discovery (CADD). In this review, we highlight recent examples of successful CADD campaigns resulting in drug candidates that directly interfere with protein-DNA interactions of several key cancer TFs, including androgen receptor (AR), ETS-related gene (ERG), MYC, thymocyte selection-associated high mobility group box protein (TOX), topoisomerase II (TOP2), and signal transducer and activator of transcription 3 (STAT3). Importantly, these findings open novel and compelling avenues for therapeutic targeting of over 1600 human TFs implicated in many conditions including and beyond cancer.
Collapse
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Anh-Tien Ton
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Michael Hsing
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
6
|
Matias-Barrios VM, Radaeva M, Song Y, Alperstein Z, Lee AR, Schmitt V, Lee J, Ban F, Xie N, Qi J, Lallous N, Gleave ME, Cherkasov A, Dong X. Discovery of New Catalytic Topoisomerase II Inhibitors for Anticancer Therapeutics. Front Oncol 2021; 10:633142. [PMID: 33598437 PMCID: PMC7883873 DOI: 10.3389/fonc.2020.633142] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/15/2020] [Indexed: 01/23/2023] Open
Abstract
Poison inhibitors of DNA topoisomerase II (TOP2) are clinically used drugs that cause cancer cell death by inducing DNA damage, which mechanism of action is also associated with serious side effects such as secondary malignancy and cardiotoxicity. In contrast, TOP2 catalytic inhibitors induce limited DNA damage, have low cytotoxicity, and are effective in suppressing cancer cell proliferation. They have been sought after to be prospective anticancer therapies. Herein the discovery of new TOP2 catalytic inhibitors is described. A new druggable pocket of TOP2 protein at its DNA binding domain was used as a docking site to virtually screen ~6 million molecules from the ZINC15 library. The lead compound, T60, was characterized to be a catalytic TOP2 inhibitor that binds TOP2 protein and disrupts TOP2 from interacting with DNA, resulting in no DNA cleavage. It has low cytotoxicity, but strongly inhibits cancer cell proliferation and xenograft growth. T60 also inhibits androgen receptor activity and prostate cancer cell growth. These results indicate that T60 is a promising candidate compound that can be further developed into new anticancer drugs.
Collapse
Affiliation(s)
- Victor M Matias-Barrios
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mariia Radaeva
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yi Song
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaccary Alperstein
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ahn R Lee
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Veronika Schmitt
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Joseph Lee
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fuqiang Ban
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ning Xie
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jianfei Qi
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Nada Lallous
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin E Gleave
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Artem Cherkasov
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xuesen Dong
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Chen L, Guo X, Hu Y, Li L, Liang G, Zhang G. Epigallocatechin-3-gallate sensitises multidrug-resistant oral carcinoma xenografts to vincristine sulfate. FEBS Open Bio 2020; 10:1403-1413. [PMID: 32475087 PMCID: PMC7327922 DOI: 10.1002/2211-5463.12905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/04/2020] [Accepted: 05/27/2020] [Indexed: 01/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a very aggressive malignancy, and 50% of patients who receive curative treatment die from the disease or related complications within 5 years. Epigallocatechin‐3‐gallate (EGCG) is the most abundant bioactive ingredient of tea polyphenols in green tea and has anticancer properties. Here, we evaluated the preclinical efficacy of EGCG combined with vincristine sulfate (VCR) on the growth, angiogenic activity and vascular endothelial growth factor (VEGF) expression in xenograft nude mice inoculated with KBV200 cells. Compared with VCR alone, the combined use of EGCG and VCR strongly inhibited tumour growth and angiogenesis (P < 0.01). VEGF mRNA and protein levels were lower in the KBV200 xenograft group treated with the combined regime (P < 0.01) than those in the VCR alone group. EGCG sensitises multidrug‐resistant OSCC to VCR, and this may occur through the inhibition of angiogenesis via VEGF down‐regulation.
Collapse
Affiliation(s)
- Li Chen
- New Drug Research & Development Center, First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.,Pharmacy School of Guangxi Medical University, Nanning, China
| | - Xianwen Guo
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ye Hu
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.,Guangxi Medical University, Nanning, China
| | - Li Li
- Pharmacy School of Guangxi Medical University, Nanning, China
| | - Gang Liang
- Pharmacy School of Guangxi Medical University, Nanning, China
| | - Guo Zhang
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
8
|
Guloglu S, Kirmaci FN, Çetinkol ÖP, Forough M, Gulkaya A. Azacyanines as Novel Topoisomerase II Alpha Inhibitors. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190628161945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Introduction:
Topoisomerase II alpha (Topo IIα) has become one of the extensively exploited
targets in chemotherapy due to its role in regulating the topological constraints of DNA during
replication and transcription. Small molecules targeting Topo IIα’s activity such as etoposide
(VP-16) and doxorubicin are extensively used in the treatment of many different types of cancer.
Objective:
Here, the effects of three small molecules, named as azacyanines, on Topo IIα have been
assessed.
Methods:
In-vitro Topoisomerase IIα drug screening kit and agarose gel imaging were used for the
assessment of Topo IIα’s activity.
Results:
Our results revealed that all the azacyanines investigated decreased the catalytic activity of
Topo IIα dramatically. More importantly, the decrease in the catalytic activity of Topo IIα in the
presence of azacyanines was higher than the presence of VP-16, which is a commercially available
chemotherapy drug. Upon further investigation, it has been observed that Azamethyl’s catalytic inhibition
of Topo IIα was concentration dependent and the catalytic activity of Topo IIα was almost
completely abolished in the presence of 100.0 μM of Azamethyl.
Conclusion:
These findings reveal the potential of azacyanines as effective Topo IIα inhibitors and
chemotherapeutic agents.
Collapse
Affiliation(s)
- Sercan Guloglu
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Fahriye Nur Kirmaci
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Özgül Persil Çetinkol
- Biochemistry Program, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Mehrdad Forough
- Department of Chemistry, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Aybuke Gulkaya
- Department of Chemistry, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| |
Collapse
|
9
|
Centenera MM, Selth LA, Ebrahimie E, Butler LM, Tilley WD. New Opportunities for Targeting the Androgen Receptor in Prostate Cancer. Cold Spring Harb Perspect Med 2018; 8:a030478. [PMID: 29530945 PMCID: PMC6280715 DOI: 10.1101/cshperspect.a030478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent genomic analyses of metastatic prostate cancer have provided important insight into adaptive changes in androgen receptor (AR) signaling that underpin resistance to androgen deprivation therapies. Novel strategies are required to circumvent these AR-mediated resistance mechanisms and thereby improve prostate cancer survival. In this review, we present a summary of AR structure and function and discuss mechanisms of AR-mediated therapy resistance that represent important areas of focus for the development of new therapies.
Collapse
Affiliation(s)
- Margaret M Centenera
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide SA 5001, Australia
| | - Luke A Selth
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Esmaeil Ebrahimie
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide SA 5001, Australia
| | - Wayne D Tilley
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide SA 5005, Australia
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
10
|
Magani F, Bray ER, Martinez MJ, Zhao N, Copello VA, Heidman L, Peacock SO, Wiley DJ, D'Urso G, Burnstein KL. Identification of an oncogenic network with prognostic and therapeutic value in prostate cancer. Mol Syst Biol 2018; 14:e8202. [PMID: 30108134 PMCID: PMC6684952 DOI: 10.15252/msb.20188202] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/11/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Identifying critical pathways governing disease progression is essential for accurate prognosis and effective therapy. We developed a broadly applicable and novel systems-level gene discovery strategy. This approach focused on constitutively active androgen receptor (AR) splice variant-driven pathways as representative of an intractable mechanism of prostate cancer (PC) therapeutic resistance. We performed a meta-analysis of human prostate samples using weighted gene co-expression network analysis combined with experimental AR variant transcriptome analyses. An AR variant-driven gene module that is upregulated during human PC progression was identified. We filtered this module by identifying genes that functionally interacted with AR variants using a high-throughput synthetic genetic array screen in Schizosaccharomyces pombe This strategy identified seven AR variant-regulated genes that also enhance AR activity and drive cancer progression. Expression of the seven genes predicted poor disease-free survival in large independent PC patient cohorts. Pharmacologic inhibition of interacting members of the gene set potently and synergistically decreased PC cell proliferation. This unbiased and novel gene discovery strategy identified a clinically relevant, oncogenic, interacting gene hub with strong prognostic and therapeutic potential in PC.
Collapse
Affiliation(s)
- Fiorella Magani
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eric R Bray
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria J Martinez
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ning Zhao
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Valeria A Copello
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laine Heidman
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephanie O Peacock
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David J Wiley
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gennaro D'Urso
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kerry L Burnstein
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center (SCCC), Miami, FL, USA
| |
Collapse
|
11
|
Gan Y, Li Y, Long Z, Lee AR, Xie N, Lovnicki JM, Tang Y, Chen X, Huang J, Dong X. Roles of Alternative RNA Splicing of the Bif-1 Gene by SRRM4 During the Development of Treatment-induced Neuroendocrine Prostate Cancer. EBioMedicine 2018; 31:267-275. [PMID: 29759485 PMCID: PMC6013970 DOI: 10.1016/j.ebiom.2018.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/12/2018] [Accepted: 05/01/2018] [Indexed: 02/02/2023] Open
Abstract
Treatment-induced neuroendocrine prostate cancer (t-NEPC) is an aggressive subtype of prostate cancer (PCa) that becomes more prevalent when hormonal therapy, chemotherapy, or radiation therapy is applied to patients with metastatic prostate adenocarcinoma (AdPC). How AdPC cells survive these anti-cancer therapies and progress into t-NEPC remains unclear. By comparing the whole transcriptomes between AdPC and t-NEPC, we identified Bif-1, an apoptosis-associated gene, which undergoes alternative RNA splicing in t-NEPC. We found that while Bif-1a is the predominant variant of the Bif-1 gene in AdPC, two neural-specific variants, Bif-1b and Bif-1c, are highly expressed in t-NEPC patients, patient derived xenografts, and cell models. The neural-specific RNA splicing factor, SRRM4, promotes Bif-1b and Bif-1c splicing, and the expression of SRRM4 in tumors is strongly associated with Bif-1b/-1c levels. Furthermore, we showed that Bif-1a is pro-apoptotic, while Bif-1b and Bif-1c are anti-apoptotic in PCa cells under camptothecin and UV light irritation treatments. Taken together, our data indicate that SRRM4 regulates alternative RNA splicing of the Bif-1 gene that enables PCa cells resistant to apoptotic stimuli under anti-cancer therapies, and may contribute to AdPC progression into t-NEPC. Alternative RNA splicing of the apoptosis-related gene, Bif-1, is associated with the development of t-NEPC. SRRM4 regulates alternative RNA splicing of the Bif-1 gene. Bif-1a in AdPC cells is pro-apoptotic, while neural Bif-1 variants, Bif-1b/-1c, enable tumor cells resistant to apoptosis.
Treatment-induced neuroendocrine prostate cancer (t-NEPC) is an aggressive subtype of castration-resistant prostate cancer. It becomes more prevalent when more potent androgen receptor inhibitors are applied to patients. However, mechanisms by which t-NEPC develops remain unclear. Here we report alternative RNA splicing of the apoptosis-related gene, Bif-1, may contribute to t-NEPC establishment. We show that the expression of neural Bif-1 variants is upregulated in t-NEPC, and confers tumor cells resistance to apoptotic stimuli. We propose that tumor cells have to first develop mechanisms to counteract therapy-induced cell death before they can undergo neuroendocrine differentiation for t-NEPC.
Collapse
Affiliation(s)
- Yu Gan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada; Department of Urology, Xiangya Hospital, Central South University, Changsha, China; Department of Urology, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yinan Li
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada.
| | - Zhi Long
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada; Department of Urology, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Ahn R Lee
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada.
| | - Ning Xie
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada.
| | - Jessica M Lovnicki
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada.
| | - Yuxin Tang
- Department of Urology, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| | - Xuesen Dong
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada.
| |
Collapse
|
12
|
Nesbitt H, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, McKenna DJ. The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate 2017; 77:1539-1547. [PMID: 28944496 DOI: 10.1002/pros.23434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/09/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND OCT1002 is a unidirectional hypoxia-activated prodrug (uHAP) OCT1002 that can target hypoxic tumor cells. Hypoxia is a common feature in prostate tumors and is known to drive disease progression and metastasis. It is, therefore, a rational therapeutic strategy to directly target hypoxic tumor cells in an attempt to improve treatment for this disease. Here we tested OCT1002 alone and in combination with standard-of-care agents in hypoxic models of castrate-resistant prostate cancer (CRPC). METHODS The effect of OCT1002 on tumor growth and vasculature was measured using murine PC3 xenograft and dorsal skin fold (DSF) window chamber models. The effects of abiraterone, docetaxel, and cabazitaxel, both singly and in combination with OCT1002, were also compared. RESULTS The hypoxia-targeting ability of OCT1002 effectively controls PC3 tumor growth. The effect was evident for at least 42 days after exposure to a single dose (30 mg/kg) and was comparable to, or better than, drugs currently used in the clinic. In DSF experiments OCT1002 caused vascular collapse in the PC3 tumors and inhibited the revascularization seen in controls. In this model OCT1002 also enhanced the anti-tumor effects of abiraterone, cabazitaxel, and docetaxel; an effect which was accompanied by a more prolonged reduction in tumor vasculature density. CONCLUSIONS These studies provide the first evidence that OCT1002 can be an effective agent in treating hypoxic, castrate-resistant prostate tumors, either singly or in combination with established chemotherapeutics for prostate cancer.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | | | - Rachel J Errington
- School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
- BioStatus Ltd, Shepshed, Leicestershire, United Kingdom
| | | | - Paul J Smith
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Stephanie R McKeown
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
13
|
Cao S, Zhan Y, Dong Y. Emerging data on androgen receptor splice variants in prostate cancer. Endocr Relat Cancer 2016; 23:T199-T210. [PMID: 27702752 PMCID: PMC5107136 DOI: 10.1530/erc-16-0298] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 12/13/2022]
Abstract
Androgen receptor splice variants are alternatively spliced variants of androgen receptor, which are C-terminally truncated and lack the canonical ligand-binding domain. Accumulating evidence has indicated a significant role of androgen receptor splice variants in mediating resistance of castration-resistant prostate cancer to current therapies and in predicting therapeutic responses. As such, there is an urgent need to target androgen receptor splicing variants for more effective treatment of castration-resistant prostate cancer. Identification of precise and critical targeting points to deactivate androgen receptor splicing variants relies on a deep understanding of how they are generated and the mechanisms of their action. In this review, we will focus on the emerging data on their generation, clinical significance and mechanisms of action as well as the therapeutic influence of these findings.
Collapse
Affiliation(s)
- Subing Cao
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Yang Zhan
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Yan Dong
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| |
Collapse
|
14
|
Bioluminescence Microscopy as a Method to Measure Single Cell Androgen Receptor Activity Heterogeneous Responses to Antiandrogens. Sci Rep 2016; 6:33968. [PMID: 27678181 PMCID: PMC5039635 DOI: 10.1038/srep33968] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022] Open
Abstract
Cancer cell heterogeneity is well-documented. Therefore, techniques to monitor single cell heterogeneous responses to treatment are needed. We developed a highly translational and quantitative bioluminescence microscopy method to measure single cell androgen receptor (AR) activity modulation by antiandrogens from fluid biopsies. We showed that this assay can detect heterogeneous cellular response to drug treatment and that the sum of single cell AR activity can mirror the response in the whole cell population. This method may thus be used to monitor heterogeneous dynamic treatment responses in cancer cells.
Collapse
|
15
|
Li H, Xie N, Chen R, Verreault M, Fazli L, Gleave ME, Barbier O, Dong X. UGT2B17 Expedites Progression of Castration-Resistant Prostate Cancers by Promoting Ligand-Independent AR Signaling. Cancer Res 2016; 76:6701-6711. [PMID: 27659047 DOI: 10.1158/0008-5472.can-16-1518] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/23/2016] [Accepted: 09/06/2016] [Indexed: 11/16/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is characterized by a shift in androgen receptor (AR) signaling from androgen-dependent to androgen (ligand)-independent. UDP-glucuronosyltransferase 2B17 (UGT2B17) is a key enzyme that maintains androgen homeostasis by catabolizing AR agonists into inactive forms. Although enhanced UGT2B17 expression by antiandrogens has been reported in androgen-dependent prostate cancer, its roles in regulating AR signaling transformation and CRPC progression remain unknown. In this study, we show that higher UGT2B17 protein expression in prostate tumors is associated with higher Gleason score, metastasis, and CRPC progression. UGT2B17 expression and activity were higher in androgen-independent compared to androgen-dependent cell lines. UGT2B17 stimulated cancer cell proliferation, invasion, and xenograft progression to CRPC after prolonged androgen deprivation. Gene microarray analysis indicated that UGT2B17 suppressed androgen-dependent AR transcriptional activity and enhanced of ligand-independent transcriptional activity at genes associated with cell mitosis. These UGT2B17 actions were mainly mediated by activation of the c-Src kinase. In CRPC tumors, UGT2B17 expression was associated positively with c-Src activation. These results indicate that UGT2B17 expedites CRPC progression by enhancing ligand-independent AR signaling to activate cell mitosis in cancer cells. Cancer Res; 76(22); 6701-11. ©2016 AACR.
Collapse
Affiliation(s)
- Haolong Li
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Ning Xie
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Ruiqi Chen
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Mélanie Verreault
- Laboratory of Molecular Pharmacology, CHU de Québec Research Centre, and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU de Québec Research Centre, and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
16
|
Tumor Repression of VCaP Xenografts by a Pyrrole-Imidazole Polyamide. PLoS One 2015; 10:e0143161. [PMID: 26571387 PMCID: PMC4646452 DOI: 10.1371/journal.pone.0143161] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/02/2015] [Indexed: 12/21/2022] Open
Abstract
Pyrrole-imidazole (Py-Im) polyamides are high affinity DNA-binding small molecules that can inhibit protein-DNA interactions. In VCaP cells, a human prostate cancer cell line overexpressing both AR and the TMPRSS2-ERG gene fusion, an androgen response element (ARE)-targeted Py-Im polyamide significantly downregulates AR driven gene expression. Polyamide exposure to VCaP cells reduced proliferation without causing DNA damage. Py-Im polyamide treatment also reduced tumor growth in a VCaP mouse xenograft model. In addition to the effects on AR regulated transcription, RNA-seq analysis revealed inhibition of topoisomerase-DNA binding as a potential mechanism that contributes to the antitumor effects of polyamides in cell culture and in xenografts. These studies support the therapeutic potential of Py-Im polyamides to target multiple aspects of transcriptional regulation in prostate cancers without genotoxic stress.
Collapse
|
17
|
Li Y, Xie N, Gleave ME, Rennie PS, Dong X. AR-v7 protein expression is regulated by protein kinase and phosphatase. Oncotarget 2015; 6:33743-54. [PMID: 26378044 PMCID: PMC4741799 DOI: 10.18632/oncotarget.5608] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/27/2015] [Indexed: 01/13/2023] Open
Abstract
Failure of androgen-targeted therapy and progression of castration-resistant prostate cancer (CRPC) are often attributed to sustained expression of the androgen receptor (AR) and its major splice variant, AR-v7. Although the new generation of anti-androgens such as enzalutamide effectively inhibits AR activity, accumulating pre-clinical and clinical evidence indicates that AR-v7 remains constitutively active in driving CRPC progression. However, molecular mechanisms which control AR-v7 protein expression remain unclear. We apply multiple prostate cancer cell models to demonstrate that enzalutamide induces differential activation of protein phosphatase-1 (PP-1) and Akt kinase depending on the gene context of cancer cells. The balance between PP-1 and Akt activation governs AR phosphorylation status and activation of the Mdm2 ubiquitin ligase. Mdm2 recognizes phosphorylated serine 213 of AR-v7, and induces AR-v7 ubiquitination and protein degradation. These findings highlight the decisive roles of PP-1 and Akt for AR-v7 protein expression and activities when AR is functionally blocked.
Collapse
Affiliation(s)
- Yinan Li
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Ning Xie
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Martin E. Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Paul S. Rennie
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Chappell WH, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Martelli AM, Cocco L, Rakus D, Gizak A, Terrian D, Steelman LS, McCubrey JA. Novel roles of androgen receptor, epidermal growth factor receptor, TP53, regulatory RNAs, NF-kappa-B, chromosomal translocations, neutrophil associated gelatinase, and matrix metalloproteinase-9 in prostate cancer and prostate cancer stem cells. Adv Biol Regul 2015; 60:64-87. [PMID: 26525204 DOI: 10.1016/j.jbior.2015.10.001] [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: 09/29/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022]
Abstract
Approximately one in six men will be diagnosed with some form of prostate cancer in their lifetime. Over 250,000 men worldwide die annually due to complications from prostate cancer. While advancements in prostate cancer screening and therapies have helped in lowering this statistic, better tests and more effective therapies are still needed. This review will summarize the novel roles of the androgen receptor (AR), epidermal growth factor receptor (EGFR), the EGFRvIII variant, TP53, long-non-coding RNAs (lncRNAs), microRNAs (miRs), NF-kappa-B, chromosomal translocations, neutrophil associated gelatinase, (NGAL), matrix metalloproteinase-9 (MMP-9), the tumor microenvironment and cancer stem cells (CSC) have on the diagnosis, development and treatment of prostate cancer.
Collapse
Affiliation(s)
- William H Chappell
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - David Terrian
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|