1
|
Al-Sawahli MM, A. El-Feky Y, Mohammed AJ, M. Mohamed N, El-Telbany R, Zuhair Alamri Z, Jameel Melebary S, Alfaifi MY, I. Elbehairi SE, M. Noreddin A, B. Abdel‑Naim A, M. Alolayan E, El-Telbany DF. Terbinafine for prostate cancer: development of coated zein nanospheres for ameliorated pro-apoptosis in PC3 cells. Nanomedicine (Lond) 2025; 20:817-833. [PMID: 40171736 PMCID: PMC11988259 DOI: 10.1080/17435889.2025.2481823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/04/2025] Open
Abstract
AIM The purpose of this study was to investigate comparatively the anticancer potential of Terbinafine loaded Dextran Sulphate coated Zein nanospheres against human prostate cancer PC3 cells to enhance the repurposing profile of terbinafine utilizing optimized nano-sized delivery systems. MATERIALS & METHODS The formula was fabricated using the thin film hydration technique. Particle size analysis, drug diffusion, and encapsulation efficiency were considered when evaluating the fabricated formula, which were developed using a Box-Behnken statistical design. RESULTS Due to the formula optimization, the mean particle size was 273.2 ± 1.98 nm, the zeta potential was -38.4 ± 2.77 mV, and the amount released after 36 h was 97.4 ± 5.7%. The formula exhibited significantly reduced IC50 in PC3 cells by around 14-fold. A higher cellular uptake was observed. The cell cycle assay results obtained suppression of the proliferation, especially in the G0/G1 and S phases. This pro-apoptotic pattern of the optimized formula was confirmed by the increased mRNA expression of CASP3 and P53 and reduced expression of CDK1, CDK7, and CDK9. Furthermore, a higher production of reactive oxygen species was achieved. CONCLUSION The optimized formula revealed enhanced pro-apoptosis in PC3 cells which support the repurposing profile of terbinafine toward prostate cancer.
Collapse
Affiliation(s)
- Majid M. Al-Sawahli
- Department of Pharmaceutics, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Yasmin A. El-Feky
- Department of Pharmaceutics, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Ahmed J. Mohammed
- Department of Clinical Laboratory Sciences, Faculty of Pharmacy, University of Kufa, Najaf, Iraq
- Department of Clinical Laboratory Sciences, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Nada M. Mohamed
- Pharmaceutical Chemistry Department, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Rania El-Telbany
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Zaenah Zuhair Alamri
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Sahar Jameel Melebary
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad Y. Alfaifi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, Saudi Arabia
| | - Serag Eldin I. Elbehairi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha, Saudi Arabia
| | - Ayman M. Noreddin
- Department of Internal Medicine, School of Medicine, University of California, Irvine, CA, USA
| | - Ashraf B. Abdel‑Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ebtesam M. Alolayan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Dalia F. El-Telbany
- Department of Pharmaceutics, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| |
Collapse
|
2
|
Deme S, Ramezani I, Coulter J, Paller C, Bressler J. Effects of hypoxia and iron on ascorbic acid-mediated cytotoxicity in prostate cancer cell lines. Toxicol Appl Pharmacol 2025; 497:117259. [PMID: 39914626 DOI: 10.1016/j.taap.2025.117259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Ascorbic acid (ASC) has long been proposed as a potential cancer co-treatment due to its specific toxicity towards cancer cells, but discrepancies between in vitro and in vivo studies suggest that external factors may modulate its cytotoxicity. Here, we investigate the impact of hypoxia and iron on the therapeutic effectiveness of ASC on prostate cancer cell lines. Hypoxia-induced increases in the EC50 of ASC in the prostate cancer cell lines PC-3, DU 145, LNCaP, and CWR22Rv1 but not in the prostate non-cancer cell lines RWPE-1 and TERT-PrECs. The synthetic androgen dihydrotestosterone did not modify ASC's effectiveness in either normoxia or hypoxia, which was tested because both early and advanced prostate cancer maintain the androgen receptor pathway. The effects of hypoxia on cytotoxicity depend on the drug. Hypoxia did not affect the EC50 for the DNA-damaging agent etoposide but decreased the sensitivity for the anti-microtubule agent paclitaxel in PC-3 and DU 145 cells. Although hypoxic cells were iron deficient, adding iron back to cells did not reverse the effects of the hypoxic atmosphere. Interestingly, the EC50 for ASC was approximately two-fold higher in iron-treated cells than non‑iron-treated cells for the PC-3 line. The higher EC50 was not observed by knocking down ferritin heavy chain mRNA. In summary, both hypoxia and iron attenuate the effectiveness of high concentrations of ASC in prostate cancer cell lines, which may affect the therapeutic benefit of ASC for prostate cancer patients.
Collapse
Affiliation(s)
- Samiksha Deme
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Baltimore, MD 21205, United States of America
| | - Ida Ramezani
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Baltimore, MD 21205, United States of America
| | - Jonathan Coulter
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States of America
| | - Channing Paller
- Department of Oncology, Johns Hopkins Medical Institute, Baltimore, MD 21205, United States of America
| | - Joseph Bressler
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Baltimore, MD 21205, United States of America.
| |
Collapse
|
3
|
Defo Deeh PB, Sathiyaseelan A, Vishven Naveen K, Wang MH. Phytochemical analysis and antioxidant potential of Mondia whitei and Guibourtia tessmannii against H 2O 2-induced cytotoxicity in PC3 cells. J Biomol Struct Dyn 2024:1-15. [PMID: 38651748 DOI: 10.1080/07391102.2024.2335299] [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: 01/24/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024]
Abstract
The management of oxidative stress-related disorders has garnered significant interest, particularly in the exploration of medicinal plants possessing potent antioxidant activities. This study was undertaken to evaluate the antioxidant activity of Mondia whitei (MW) and Guibourtia tessmannii (GT) against H2O2-induced cytotoxicity in PC3 cells. The phytochemical composition of MW and GT was determined by GC-MS analysis. Total phenolic (TP) and total flavonoid (TF) contents were quantified by Folin Ciocalteu and AlCl3 methods, respectively. The antioxidant potential of the extracts was determined using the DPPH and ABTS+ radicals scavenging method, as well as cupric and ferric reducing capacity assay. Moreover, all phytocompounds were docked against acetylcholinesterase (AChE) and glutathione S-transferase (GST) using ArgusLab, and results were analyzed using the BIOVIA Discovery Studio Visualizer 2021 client. MW and GT comprised 20 and 22 compounds, respectively. GT exhibited higher TP and TF contents (210.70 ± 12.7; 12.61 ± 1.3 GAE/g DW) compared to MW (132.59 ± 12.59; 5.53 ± 1.3 mg of GAE/g DW). Both MW and GT demonstrated substantial antioxidant activity, with GT proving to be more effective in preventing H2O2-induced cytotoxicity. For instance, MW and GT significantly (p < .001) increased the DPPH, ABTS+, and cupric activity, compared with the H2O2 group. All compounds identified in MW and GT exhibited a strong binding affinity against AChE and GST. Drug likeness and toxicity of all phytocompounds were under the acceptable norms of Lipinski's rule. In conclusion, these plants could be effective candidates for the management/treatment of oxidative stress-related disorders.
Collapse
Affiliation(s)
- Patrick Brice Defo Deeh
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | | | - Kumar Vishven Naveen
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
4
|
Yang J, Xin C, Yin G, Li J. Taraxasterol suppresses the proliferation and tumor growth of androgen-independent prostate cancer cells through the FGFR2-PI3K/AKT signaling pathway. Sci Rep 2023; 13:13072. [PMID: 37567936 PMCID: PMC10421874 DOI: 10.1038/s41598-023-40344-w] [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: 01/03/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate cancer (PCa) is prevalent among older men and difficult to survive after metastasis. It is urgent to find new drugs and treatments. Several studies show that taraxasterol (TAX) has important anti-inflammatory, anti-oxidative and anti-tumor effects. However, the function and mechanisms of TAX in PCa remain unclear. Here, we found that TAX could significantly suppress the viability and growth of androgen-independent PCa cells and down-regulate the expression of c-Myc and cyclin D1 in vitro. Mechanistically, PI3K/AKT signaling pathway was weakened and the expression of FGFR2 was reduced after TAX treatment in androgen-independent PCa cells. Moreover, TAX evidently inhibited the tumor growth in nude mice and the expression of c-Myc, cyclin D1, p-AKT and FGFR2 were down-regulated in xenograft tumor. These results indicate that TAX suppresses the proliferation of androgen-independent PCa cells via inhibiting the activation of PI3K/AKT signaling pathway and the expression of FGFR2, which means TAX may be a novel anti-tumor agent for later PCa treatment.
Collapse
Affiliation(s)
- Jinqiu Yang
- School of Clinical Medicine, Dali University, Dali, 671013, Yunnan, China
| | - Chulin Xin
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China
| | - Guangfen Yin
- The First Affiliated Hospital of Dali University, Dali, 671013, Yunnan, China
| | - Juan Li
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China.
| |
Collapse
|
5
|
Abdelwahed KS, Siddique AB, Ebrahim HY, Qusa MH, Mudhish EA, Rad AH, Zerfaoui M, Abd Elmageed ZY, El Sayed KA. Pseurotin A Validation as a Metastatic Castration-Resistant Prostate Cancer Recurrence-Suppressing Lead via PCSK9-LDLR Axis Modulation. Mar Drugs 2023; 21:215. [PMID: 37103355 PMCID: PMC10144979 DOI: 10.3390/md21040215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) cells can de novo biosynthesize their own cholesterol and overexpress proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 proved to contribute to mCRPC cell motility since PCSK9 knockdown (KD) in mCRPC CWR-R1ca cells led to notable reductions in cell migration and colony formation. Human tissue microarray results proved a higher immunohistoscore in patients ≥ 65 years old, and PCSK9 proved to be expressed higher at an early Gleason score of ≤7. The fermentation product pseurotin A (PS) suppressed PCSK9 expression, protein-protein interactions with LDLR, and breast and prostate cancer recurrences. PS suppressed migration and colony formation of the CWR-R1ca cells. The progression and metastasis of the CWR-R1ca-Luc cells subcutaneously (sc) xenografted into male nude mice fed a high-fat diet (HFD, 11% fat content) showed nearly 2-fold tumor volume, metastasis, serum cholesterol, low-density lipoprotein cholesterol (LDL-C), prostate-specific antigen (PSA), and PCSK9 levels versus mice fed a regular chow diet. Daily oral PS 10 mg/kg treatments prevented the locoregional and distant tumor recurrence of CWR-R1ca-Luc engrafted into nude mice after primary tumor surgical excision. PS-treated mice showed a significant reduction in serum cholesterol, LDL-C, PCSK9, and PSA levels. These results comprehensively validate PS as an mCRPC recurrence-suppressive lead by modulating the PCSK9-LDLR axis.
Collapse
Affiliation(s)
- Khaldoun S. Abdelwahed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Abu Bakar Siddique
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Hassan Y. Ebrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Mohammed H. Qusa
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Ethar A. Mudhish
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Ashkan H. Rad
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Mourad Zerfaoui
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Zakaria Y. Abd Elmageed
- Department of Biomedical Sciences, Discipline of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana at Monroe, Monroe, LA 71203, USA
| | - Khalid A. El Sayed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
6
|
Sena LA, Kumar R, Sanin DE, Thompson EA, Rosen DM, Dalrymple SL, Antony L, Yang Y, Gomes-Alexandre C, Hicks JL, Jones T, Bowers KA, Eskra JN, Meyers J, Gupta A, Skaist A, Yegnasubramanian S, Luo J, Brennen WN, Kachhap SK, Antonarakis ES, De Marzo AM, Isaacs JT, Markowski MC, Denmeade SR. Androgen receptor activity in prostate cancer dictates efficacy of bipolar androgen therapy through MYC. J Clin Invest 2022; 132:e162396. [PMID: 36194476 PMCID: PMC9711876 DOI: 10.1172/jci162396] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Testosterone is the canonical growth factor of prostate cancer but can paradoxically suppress its growth when present at supraphysiological levels. We have previously demonstrated that the cyclical administration of supraphysiological androgen (SPA), termed bipolar androgen therapy (BAT), can result in tumor regression and clinical benefit for patients with castration-resistant prostate cancer. However, predictors and mechanisms of response and resistance have been ill defined. Here, we show that growth inhibition of prostate cancer models by SPA required high androgen receptor (AR) activity and were driven in part by downregulation of MYC. Using matched sequential patient biopsies, we show that high pretreatment AR activity predicted downregulation of MYC, improved clinical response, and prolonged progression-free and overall survival for patients on BAT. BAT induced strong downregulation of AR in all patients, which is shown to be a primary mechanism of acquired resistance to SPA. Acquired resistance was overcome by alternating SPA with the AR inhibitor enzalutamide, which induced adaptive upregulation of AR and resensitized prostate cancer to SPA. This work identifies high AR activity as a predictive biomarker of response to BAT and supports a treatment paradigm for prostate cancer involving alternating between AR inhibition and activation.
Collapse
|
7
|
Peinetti N, Bilusic M, Burnstein KL. Is androgen receptor activity in metastatic prostate cancer a good biomarker for bipolar androgen therapy? J Clin Invest 2022; 132:e165357. [PMID: 36453547 PMCID: PMC9711867 DOI: 10.1172/jci165357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the longstanding treatment for advanced prostate cancer (PC) because androgen receptor (AR) is the key therapeutic vulnerability for this disease. Bipolar androgen therapy (BAT) - the rapid cycling of supraphysiologic androgen (SPA) and low serum testosterone levels - is an alternative concept, but not all patients respond and acquired resistance can occur. In this issue of the JCI, Sena et al. developed a gene signature indicative of high AR activity to predict patient response to BAT, including a decline in both serum prostate-specific antigen (PSA) and tumor volume. Preclinical models showed that AR-mediated suppression of MYC, known to drive PC, was associated with decreased cell growth following SPA treatment. Because BAT eventually leads to resistance, the authors tested cycling between SPA and AR antagonism in a patient-derived xenograft and observed a delay in tumor growth. These findings represent a major step toward the informed use of BAT for advanced PC.
Collapse
Affiliation(s)
- Nahuel Peinetti
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Marijo Bilusic
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kerry L. Burnstein
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
- Department of Veterans Affairs, Miami, Florida, USA
| |
Collapse
|
8
|
Sienkiewicz K, Yang C, Paschal BM, Ratan A. Genomic analyses of the metastasis-derived prostate cancer cell lines LNCaP, VCaP, and PC3-AR. Prostate 2022; 82:442-451. [PMID: 34951700 PMCID: PMC8792310 DOI: 10.1002/pros.24290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND The lymph node metastasis-derived LNCaP, the bone metastasis-derived PC3 (skull), and VCaP (vertebral) cell lines are widely used as preclinical models of human prostate cancer (CaP) and have been described in more than 19,000 publications. Here, we report on short-read whole-genome sequencing and genomic analyses of LNCaP, VCaP, and PC3 cells stably transduced with WT AR (PC3-AR). METHODS LNCaP, VCaP, and PC3-AR cell lines were sequenced to an average depth of more than 30-fold using Illumina short-read sequencing. Using various computational methods, we identified and compared the single-nucleotide variants, copy-number profiles, and the structural variants observed in the three cell lines. RESULTS LNCaP cells are composed of multiple subpopulations, which results in nonintegral copy number states and a high mutational load when the data is analyzed in bulk. All three cell lines contain pathogenic mutations and homozygous deletions in genes involved in DNA mismatch repair, along with deleterious mutations in cell-cycle, Wnt signaling, and other critical cellular processes. PC3-AR cells have a truncating mutation in TP53 and do not express the p53 protein. The VCaP cells contain a homozygous gain-of-function mutation in TP53 (p.R248W) that promotes cancer invasion, metastasis, and progression and has also been observed in prostate adenocarcinomas. In addition, we detect the signatures of chromothripsis of the q arms of chromosome 5 in both PC3-AR and VCaP cells, strengthening the association of TP53 inactivation with chromothripsis reported in other systems. CONCLUSIONS Our work provides a resource for genetic, genomic, and biological studies employing these commonly-used prostate cancer cell lines.
Collapse
Affiliation(s)
| | - Chunsong Yang
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Bryce M. Paschal
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Virginia, USA
- Department of Public Health Sciences, University of Virginia, Virginia, USA
| |
Collapse
|
9
|
Leach DA, Fernandes RC, Bevan CL. Cellular specificity of androgen receptor, coregulators, and pioneer factors in prostate cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2022; 2:R112-R131. [PMID: 37435460 PMCID: PMC10259329 DOI: 10.1530/eo-22-0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 07/13/2023]
Abstract
Androgen signalling, through the transcription factor androgen receptor (AR), is vital to all stages of prostate development and most prostate cancer progression. AR signalling controls differentiation, morphogenesis, and function of the prostate. It also drives proliferation and survival in prostate cancer cells as the tumour progresses; given this importance, it is the main therapeutic target for disseminated disease. AR is also essential in the surrounding stroma, for the embryonic development of the prostate and controlling epithelial glandular development. Stromal AR is also important in cancer initiation, regulating paracrine factors that excite cancer cell proliferation, but lower stromal AR expression correlates with shorter time to progression/worse outcomes. The profile of AR target genes is different between benign and cancerous epithelial cells, between castrate-resistant prostate cancer cells and treatment-naïve cancer cells, between metastatic and primary cancer cells, and between epithelial cells and fibroblasts. This is also true of AR DNA-binding profiles. Potentially regulating the cellular specificity of AR binding and action are pioneer factors and coregulators, which control and influence the ability of AR to bind to chromatin and regulate gene expression. The expression of these factors differs between benign and cancerous cells, as well as throughout disease progression. The expression profile is also different between fibroblast and mesenchymal cell types. The functional importance of coregulators and pioneer factors in androgen signalling makes them attractive therapeutic targets, but given the contextual expression of these factors, it is essential to understand their roles in different cancerous and cell-lineage states.
Collapse
Affiliation(s)
- Damien A Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Rayzel C Fernandes
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
10
|
Avsar Abdik E, Abdik H, Turan D, Sahin F, Berger MR, Kaleagasioglu F. Dual Akt and Bcl-2 inhibition induces cell-type specific modulation of apoptotic and autophagic signaling in castration resistant prostate cancer cell lines. Mol Biol Rep 2021; 48:7755-7765. [PMID: 34647221 DOI: 10.1007/s11033-021-06786-z] [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: 07/06/2021] [Accepted: 09/08/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cancer cell survival depends on the cross-regulation between apoptosis and autophagy which share common signaling pathways including PI3K/Akt/mTOR and Bcl-2. The aim of this study was to elucidate the modulation patterns between apoptosis and autophagy following dual inhibition by Akt inhibitor erufosine and Bcl-2 inhibitor ABT-737 in castration-resistant prostate cancer (CRPC) cell lines, PC-3 (Bax+) and DU-145 (Bax-). METHODS AND RESULTS Cell cycle progression, apoptotic and autophagic signaling were examined by flow cytometry, multi-caspase assay, Hoechst staining, acridine orange staining of acidic vesicular organelles (AVOs), qRT-PCR and Western Blot. Dual inhibition increased G2/M arrest in PC-3 and DU-145, but not in the healthy prostate epithelium cells, PNT-1A. Only in PC-3, dual inhibition induced synergistic apoptotic and additive autophagic effects. In DU-145 and PNT-1A cells, ABT-737 did not display any remarkable effect on multicaspase activity and erufosine and ABT-737, neither alone nor in combination induced AVOs. By dual inhibition, AKT, BCL-2 and NF-κB gene expressions were downregulated in PC-3, both ATG-5 and BECLIN-1 gene expressions were upregulated in DU-145 but Beclin-1 protein expression was substantially reduced in both CRPC cells. Dual inhibition-induced synergistic multicaspase activation in PC-3 degrades and disrupts autophagic activity of Beclin-1, enhancing caspase-dependent apoptosis. However, in DU-145, following dual inhibition, rate of multicaspase induction and apoptosis are lower but autophagy is completely abolished despite markedly increased BECLIN-1 gene expression. CONCLUSION In conclusion, antineoplastic drug combinations may display cell-type specific modulation of apoptotic and autophagic signaling and lack of protective autophagy may not necessarily indicate increased chemotherapeutic sensitivity in heterogenous tumor subpopulations.
Collapse
Affiliation(s)
- Ezgi Avsar Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Hüseyin Abdik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkey
| | - Duygu Turan
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Koç University, Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
| | - Ferda Kaleagasioglu
- Department of Pharmacology and Clinical Pharmacology, Istinye University Faculty of Medicine, Topkapı Campus, Maltepe Neighbourhood, Teyyareci Sami St., No. 3, Zeytinburnu, Istanbul, Turkey.
| |
Collapse
|
11
|
Zhong S, Jeong JH, Huang C, Chen X, Dickinson SI, Dhillon J, Yang L, Luo JL. Targeting INMT and interrupting its methylation pathway for the treatment of castration resistant prostate cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:307. [PMID: 34587977 PMCID: PMC8482636 DOI: 10.1186/s13046-021-02109-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/19/2021] [Indexed: 02/07/2023]
Abstract
Background Castration-resistant prostate cancer (CRPC) is associated with a very poor prognosis, and the treatment of which remains a serious clinical challenge. Methods RNA-seq, qPCR, western blot and immunohistochemistry were employed to identify and confirm the high expression of indolethylamine N-methyltransferase (INMT) in CRPC and the clinical relevance. Chip assay was used to identify Histone-Lysine N-Methyltransferase (SMYD3) as a major epigenetic regulator of INMT. LC-MS/MS were used to identify new substrates of INMT methylation in CRPC tissues. Gene knockdown/overexpression, MTT and mouse cancer models were used to examine the role of INMT as well as the anticancer efficacy of INMT inhibitor N,N-dimethyltryptamine (DMT), the SMYD3 inhibitor BCl-12, the selenium compounds methaneseleninic acid (MSA) and Se-(Methyl)selenocysteine hydrochloride (MSC), and the newly identified endogenous INMT substrate Bis(7)-tacrine. Results We found that the expression of INMT was highly increased in CRPC and was correlated with poor prognosis of clinical prostate cancer (PCa). INMT promoted PCa castration resistance via detoxification of anticancer metabolites. Knockdown of INMT or treatment with INMT inhibitor N,N-dimethyltryptamine (DMT) significantly suppressed CRPC development. Histone-Lysine N-Methyltransferase SMYD3 was a major epigenetic regulator of INMT expression, treatment with SMYD3 inhibitor BCl-121 suppressed INMT expression and inhibits CRPC development. Importantly, INMT knockdown significantly increased the anticancer effect of the exogenous selenium compounds methaneseleninic acid (MSA) and Se-(Methyl)selenocysteine hydrochloride (MSC) as well as the endogenous metabolite Bis(7)-tacrine. Conclusions Our study suggests that INMT drives PCa castration resistance through detoxification of anticancer metabolites, targeting INMT or its regulator SMYD3 or/and its methylation metabolites represents an effective therapeutic avenue for CRPC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02109-z.
Collapse
Affiliation(s)
- Shangwei Zhong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Ji-Hak Jeong
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA.,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Changhao Huang
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Xueyan Chen
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | | | - Jasreman Dhillon
- Department of Pathology, Moffitt Cancer Center, 2902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Li Yang
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA. .,Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.
| | - Jun-Li Luo
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA.
| |
Collapse
|
12
|
Jamroze A, Chatta G, Tang DG. Androgen receptor (AR) heterogeneity in prostate cancer and therapy resistance. Cancer Lett 2021; 518:1-9. [PMID: 34118355 DOI: 10.1016/j.canlet.2021.06.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/16/2021] [Accepted: 06/06/2021] [Indexed: 12/30/2022]
Abstract
Androgen receptor (AR), a ligand-dependent nuclear transcription factor and a member of steroid hormone receptor family, plays an important role in prostate organogenesis by regulating epithelial differentiation and restricting cell proliferation. Although rarely mutated or amplified in treatment-naïve prostate cancer (PCa), AR signaling drives tumor growth and as a result, therapies that aim to inhibit AR signaling, called ARSIs (AR signaling inhibitors), have been in clinical use for >70 years. Unfortunately, the clinical efficacy of ARSIs is short-lived and the majority of treated patients develop castration-resistant PCa (CRPC). Numerous molecular mechanisms have been proposed for castration resistance; however, the cellular basis for CRPC emergence has remained obscure. One under-appreciated cellular mechanism for CRPC development is the AR heterogeneity that pre-exists in treatment-naive primary tumors, i.e., although most PCa cells express AR (i.e., AR+), there is always a population of PCa cells that express no/low AR (i.e., AR-/lo). Importantly, this AR heterogeneity becomes accentuated during ARSI treatment and highly prominent in established CRPC. Here, we provide a succinct summary of AR heterogeneity across the PCa continuum and discuss its impact on PCa response to treatments. While AR+ PCa cells/clones exhibit exquisite sensitivities to ARSIs, AR-/lo PCa cells/clones, which are greatly enriched in stem cell signaling pathways, display de novo resistance to ARSIs. Finally, we offer several potential combinatorial strategies, e.g., ARSIs with stem cell targeting therapeutics, to co-target both AR+ and AR-/lo PCa cells and metastatic clones.
Collapse
Affiliation(s)
- Anmbreen Jamroze
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, NY, 14263, USA.
| |
Collapse
|
13
|
Transcriptomic Analysis of LNCaP Tumor Xenograft to Elucidate the Components and Mechanisms Contributed by Tumor Environment as Targets for Dietary Prostate Cancer Prevention Studies. Nutrients 2021; 13:nu13031000. [PMID: 33808801 PMCID: PMC8003580 DOI: 10.3390/nu13031000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/24/2022] Open
Abstract
LNCaP athymic xenograft model has been widely used to allow researchers to examine the effects and mechanisms of experimental treatments such as diet and diet-derived cancer preventive and therapeutic compounds on prostate cancer. However, the biological characteristics of human LNCaP cells before/after implanting in athymic mouse and its relevance to clinical human prostate outcomes remain unclear and may dictate interpretation of biological efficacies/mechanisms of diet/diet-derived experimental treatments. In this study, transcriptome profiles and pathways of human prostate LNCaP cells before (in vitro) and after (in vivo) implanting into xenograft mouse were compared using RNA-sequencing technology (RNA-seq) followed by bioinformatic analysis. A shift from androgen-responsive to androgen nonresponsive status was observed when comparing LNCaP xenograft tumor to culture cells. Androgen receptor and aryl-hydrocarbon pathway were found to be inhibited and interleukin-1 (IL-1) mediated pathways contributed to these changes. Coupled with in vitro experiments modeling for androgen exposure, cell-matrix interaction, inflammation, and hypoxia, we identified specific mechanisms that may contribute to the observed changes in genes and pathways. Our results provide critical baseline transcriptomic information for a tumor xenograft model and the tumor environments that might be associated with regulating the progression of the xenograft tumor, which may influence interpretation of diet/diet-derived experimental treatments.
Collapse
|
14
|
Wang J, Xu LF, Liu C, Huang T, Liang CZ, Fan YD. Identifying the role of apolipoprotein A-I in prostate cancer. Asian J Androl 2021; 23:400-408. [PMID: 33586698 PMCID: PMC8269822 DOI: 10.4103/aja.aja_92_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although localized prostate cancer (PCa) can be cured by prostatectomy and radiotherapy, the development of effective therapeutic approaches for advanced prostate cancer, including castration-resistant PCa (CRPC) and neuroendocrine PCa (NEPC), is lagging far behind. Identifying a novel prognostic and diagnostic biomarker for early diagnosis and intervention is an urgent clinical need. Here, we report that apolipoprotein A-I (ApoA-I), the major component of high-density lipoprotein (HDL), is upregulated in PCa based on both bioinformatics and experimental evidence. The fact that advanced PCa shows strong ApoA-I expression reflects its potential role in driving therapeutic resistance and disease progression by reprogramming the lipid metabolic network of tumor cells. Molecularly, ApoA-I is regulated by MYC, a frequently amplified oncogene in late-stage PCa. Altogether, our findings have revealed a novel indicator to predict prognosis and recurrence, which would benefit patients who are prone to progress to metastasis or even NEPC, which is the lethal subtype of PCa.
Collapse
Affiliation(s)
- Jing Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.,Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Ling-Fan Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Cheng Liu
- Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Tao Huang
- Department of Urologic Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yi-Dong Fan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
15
|
Del Bubba M, Di Serio C, Renai L, Scordo CVA, Checchini L, Ungar A, Tarantini F, Bartoletti R. Vaccinium myrtillus
L. extract and its native polyphenol‐recombined mixture have anti‐proliferative and pro‐apoptotic effects on human prostate cancer cell lines. Phytother Res 2020; 35:1089-1098. [DOI: 10.1002/ptr.6879] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/17/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022]
Affiliation(s)
| | - Claudia Di Serio
- Department of Clinical and Experimental Medicine University of Florence Florence Italy
| | - Lapo Renai
- Department of Chemistry University of Florence Florence Italy
| | | | | | - Andrea Ungar
- Department of Clinical and Experimental Medicine University of Florence Florence Italy
| | - Francesca Tarantini
- Department of Clinical and Experimental Medicine University of Florence Florence Italy
| | - Riccardo Bartoletti
- Department of Translational Research and New Technologies in Medicine and Surgery University of Pisa Pisa Italy
| |
Collapse
|
16
|
Wróbel TM, Rogova O, Andersen KL, Yadav R, Brixius-Anderko S, Scott EE, Olsen L, Jørgensen FS, Björkling F. Discovery of Novel Non-Steroidal Cytochrome P450 17A1 Inhibitors as Potential Prostate Cancer Agents. Int J Mol Sci 2020; 21:ijms21144868. [PMID: 32660148 PMCID: PMC7402352 DOI: 10.3390/ijms21144868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The current study presents the design, synthesis, and evaluation of novel cytochrome P450 17A1 (CYP17A1) ligands. CYP17A1 is a key enzyme in the steroidogenic pathway that produces androgens among other steroids, and it is implicated in prostate cancer. The obtained compounds are potent enzyme inhibitors (sub µM) with antiproliferative activity in prostate cancer cell lines. The binding mode of these compounds is also discussed.
Collapse
Affiliation(s)
- Tomasz M. Wróbel
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
- Correspondence: ; Tel.: +48-814-487-273
| | - Oksana Rogova
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| | - Kasper L. Andersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark;
| | - Rahul Yadav
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
| | - Simone Brixius-Anderko
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
| | - Emily E. Scott
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
- Department of Pharmacology, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA
| | - Lars Olsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
- Protein Engineering, Novozymes A/S, Krogshøjvej 36, DK-2880 Bagsvaerd, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| |
Collapse
|
17
|
Chaves FM, Pavan ICB, da Silva LGS, de Freitas LB, Rostagno MA, Antunes AEC, Bezerra RMN, Simabuco FM. Pomegranate Juice and Peel Extracts are Able to Inhibit Proliferation, Migration and Colony Formation of Prostate Cancer Cell Lines and Modulate the Akt/mTOR/S6K Signaling Pathway. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2020; 75:54-62. [PMID: 31838616 DOI: 10.1007/s11130-019-00776-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pomegranate (Punica granatum) is known to contain polyphenols with many potential health benefits, including anti-tumoral, anti-inflammatory, and anti-microbial properties. It has been used in popular medicine for cancer treatment, which still represents the major cause of cancer-related deaths in men worldwide. Importantly, pomegranate peels are valuable by-products of the food industry that are rich in polyphenols. Here we report a comparison between juice and peel aqueous extracts in prostate cancer DU-145 and PC-3 cell lines. Both extracts were able to inhibit the proliferation, migration and colony formation of those cells, although peel extracts presented more robust effects compared to juice. Besides, the growth-related mTOR/S6K signaling pathway presented strong inhibition after pomegranate extracts treatment. This study presents evidence that both juice and isolated peel extracts from promegate fruit have important anti-cancer effects against prostate cancer cells, modulating the mTOR/S6K signaling pathway.
Collapse
Affiliation(s)
- Fernanda Machado Chaves
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
- Dairy, Probiotics and Prebiotics Laboratory (LLPP), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Lidia Broglio de Freitas
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mauricio Ariel Rostagno
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Adriane Elisabete Costa Antunes
- Dairy, Probiotics and Prebiotics Laboratory (LLPP), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rosângela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil.
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| |
Collapse
|
18
|
Lai HW, Sasaki R, Usuki S, Nakajima M, Tanaka T, Ogura SI. Novel strategy to increase specificity of ALA-Induced PpIX accumulation through inhibition of transporters involved in ALA uptake. Photodiagnosis Photodyn Ther 2019; 27:327-335. [DOI: 10.1016/j.pdpdt.2019.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/20/2019] [Accepted: 06/24/2019] [Indexed: 01/10/2023]
|
19
|
Tsang M, Gantchev J, Netchiporouk E, Moreau L, Ghazawi FM, Glassman S, Sasseville D, Litvinov IV. A study of meiomitosis and novel pathways of genomic instability in cutaneous T-cell lymphomas (CTCL). Oncotarget 2018; 9:37647-37661. [PMID: 30701021 PMCID: PMC6340880 DOI: 10.18632/oncotarget.26479] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
Genomic instability is a hallmark of cancer and an enabling factor for genetic alterations that drive cancer development and progression. The clashing of mitosis and aberrantly expressed meiosis machineries, which may contribute to genomic instability, has been coined cancer “meiomitosis”. LINE-1 retrotransposition, a process active in germ cells, acts outside of the meiotic machinery to create DNA double strand breaks (DNA DSBs) and has played an important role in the evolution of the human genome. We have previously demonstrated that in CTCL several cancer testis/meiotic genes are expressed. Furthermore, this cancer exhibits extensive and ongoing chromosomal/microsatellite instability. In this study we analyzed immortalized patient-derived cells and primary CTCL patient samples using RT-PCR, western blotting and confocal microscopy and found that proteins critically involved in meiosis and LINE-1 retrotransposition are expressed and are associated with chromosomal instability and DNA DSB formation. Using cell cycle synchronization, we show G1/S phase-transition-specific expression of meiosis proteins. Using the Alu retrotransposition assay, we demonstrate the functional activity of LINE-1 retrotransposon in CTCL. Histone acetyltransferase inhibition results in downregulation of the ectopic germ cell programs and concomitant decrease in DNA DSBs foci formation. Notably, LINE-1 and meiosis genes were expressed across a panel of other solid tumor cell lines. Taken together, our results indicate that malignant cells in culture undergo “cancer meiomitosis” rather than the classic mitosis division. The ectopic expression of meiosis genes and reactivation of LINE-1 may be contributing to genomic instability and represent novel targets for immunotherapy in this and other cancers.
Collapse
Affiliation(s)
- Matthew Tsang
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Jennifer Gantchev
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Elena Netchiporouk
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Linda Moreau
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Feras M Ghazawi
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Steven Glassman
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada
| | - Denis Sasseville
- Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Ivan V Litvinov
- Division of Dermatology, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada.,Division of Dermatology, McGill University, Montréal, Québec H4A 3J1, Canada
| |
Collapse
|
20
|
Geng H, Xue C, Mendonca J, Sun XX, Liu Q, Reardon PN, Chen Y, Qian K, Hua V, Chen A, Pan F, Yuan J, Dang S, Beer TM, Dai MS, Kachhap SK, Qian DZ. Interplay between hypoxia and androgen controls a metabolic switch conferring resistance to androgen/AR-targeted therapy. Nat Commun 2018; 9:4972. [PMID: 30478344 PMCID: PMC6255907 DOI: 10.1038/s41467-018-07411-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Despite recent advances, the efficacy of androgen/androgen receptor (AR)-targeted therapy remains limited for many patients with metastatic prostate cancer. This is in part because prostate cancers adaptively switch to the androgen/AR-independent pathway for survival and growth, thereby conferring therapy resistance. Tumor hypoxia is considered as a major cause of treatment resistance. However, the exact mechanism is largely unclear. Here we report that chronic-androgen deprivation therapy (ADT) in the condition of hypoxia induces adaptive androgen/AR-independence, and therefore confers resistance to androgen/AR-targeted therapy, e.g., enzalutamide. Mechanistically, this is mediated by glucose-6-phosphate isomerase (GPI), which is transcriptionally repressed by AR in hypoxia, but restored and increased by AR inhibition. In turn, GPI maintains glucose metabolism and energy homeostasis in hypoxia by redirecting the glucose flux from androgen/AR-dependent pentose phosphate pathway (PPP) to hypoxia-induced glycolysis pathway, thereby reducing the growth inhibitory effect of enzalutamide. Inhibiting GPI overcomes the therapy resistance in hypoxia in vitro and increases enzalutamide efficacy in vivo.
Collapse
Affiliation(s)
- Hao Geng
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Changhui Xue
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Janet Mendonca
- Johns Hopkins Kimmel Cancer Center, 401 N Broadway, Baltimore, MD, 21287, USA
| | - Xiao-Xin Sun
- Department of Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Qiong Liu
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Patrick N Reardon
- NMR Core facility, Oregon State University, Corvallis, OR, 97331, USA
| | - Yingxiao Chen
- Department of Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Kendrick Qian
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Vivian Hua
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Alice Chen
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Freddy Pan
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Julia Yuan
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Sang Dang
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Tomasz M Beer
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Division of Hematology & Medical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Mu-Shui Dai
- Department of Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Sushant K Kachhap
- Johns Hopkins Kimmel Cancer Center, 401 N Broadway, Baltimore, MD, 21287, USA
| | - David Z Qian
- OHSU Knight Cancer Institute, Prostate Cancer Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
- Division of Hematology & Medical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
21
|
Lam HM, Corey E. Supraphysiological Testosterone Therapy as Treatment for Castration-Resistant Prostate Cancer. Front Oncol 2018; 8:167. [PMID: 29872642 PMCID: PMC5972313 DOI: 10.3389/fonc.2018.00167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/01/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
22
|
Park SW, Kim JH, Lee HJ, Shin DH, Lee SD, Yoon S. The Expression of Androgen Receptor and Its Variants in Human Prostate Cancer Tissue according to Disease Status, and Its Prognostic Significance. World J Mens Health 2018; 37:68-77. [PMID: 29756415 PMCID: PMC6305860 DOI: 10.5534/wjmh.180003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 11/15/2022] Open
Abstract
Purpose To evaluate changes in the expression of androgen receptor (AR) and its variants (ARVs) in human prostate cancer (PCa) tissues according to disease status, and its prognostic significance following radical prostatectomy (RP). Materials and Methods A total of 282 PCa cases were evaluated, which included 252 localized PCa, 8 metastatic castration resistant prostate cancer (CRPC), and 22 benign prostatic hyperplasia (BPH) cases. Samples were collected from patients who underwent RP or transurethral resection and were stored in ethically approved tissue banks. Quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry were performed for AR and ARVs. Each tissue was confirmed as cancerous (greater than 80%) using hematoxylin and eosin staining. AR and ARVs expression was compared according to disease status. The biochemical recurrence free survival (BCRFS) rates in men with localized PCa was analyzed according to AR and ARV7 expression using the Kaplan-Meier curve. Results Only 58 of the 252 localized PCa were included in the analysis because of insufficient cancer tissue. AR and ARV7 mRNA expression was higher in the CRPC tissue than in the localized PCa tissue (p=0.025, p=0.002, respectively). In localized PCa tissue, high AR mRNA and protein level was associated with a low BCRFS rate (log-ranked, p=0.019, p<0.001, respectively). Conclusions Overall AR and ARV7 mRNA expression levels were increased in CRPC tissues compared to localized PCa and BPH tissues. High AR protein and mRNA expression in the tumor tissue may be considered a predictive factor of BCRFS following RP.
Collapse
Affiliation(s)
- Sung Woo Park
- Department of Urology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jung Hee Kim
- Department of Urology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Hyun Jung Lee
- Department of Pathology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Dong Hoon Shin
- Department of Pathology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sang Don Lee
- Department of Urology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sik Yoon
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.
| |
Collapse
|
23
|
Androgen receptor differentially regulates the proliferation of prostatic epithelial cells in vitro and in vivo. Oncotarget 2018; 7:70404-70419. [PMID: 27611945 PMCID: PMC5342561 DOI: 10.18632/oncotarget.11879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/24/2016] [Indexed: 01/13/2023] Open
Abstract
Androgens regulate the proliferation and differentiation of prostatic epithelial cells, including prostate cancer (PCa) cells in a context-dependent manner. Androgens and androgen receptor (AR) do not invariably promote cell proliferation; in the normal adult, endogenous stromal and epithelial AR activation maintains differentiation and inhibits organ growth. In the current study, we report that activation of AR differentially regulates the proliferation of human prostate epithelial progenitor cells, NHPrE1, in vitro and in vivo. Inducing AR signaling in NHPrE1 cells suppressed cell proliferation in vitro, concomitant with a reduction in MYC expression. However, ectopic expression of AR in vivo stimulated cell proliferation and induced development of invasive PCa in tissue recombinants consisting of NHPrE1/AR cells and rat urogenital mesenchymal (UGM) cells, engrafted under renal capsule of adult male athymic mice. Expression of MYC increased in the NHPrE1/AR recombinant tissues, in contrast to the reduction seen in vitro. The inhibitory effect of AR signaling on cell proliferation in vitro were reduced by co-culturing NHPrE1/AR epithelial cells with prostatic stromal cells. In conclusion, these studies revealed that AR signaling differentially regulates proliferation of human prostatic epithelia cells in vitro and in vivo through mechanisms involving stromal/epithelial interactions.
Collapse
|
24
|
Wang J, Zhang H, Zhang X, Wang P, Wang H, Huang F, Zhou C, Zhou J, Li S. PC-1 works in conjunction with E3 ligase CHIP to regulate androgen receptor stability and activity. Oncotarget 2018; 7:81377-81388. [PMID: 27835608 PMCID: PMC5348399 DOI: 10.18632/oncotarget.13230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 11/01/2016] [Indexed: 01/26/2023] Open
Abstract
The androgen receptor (AR) is not only a ligand-dependent transcription factor, but also functions as a licensing factor, a component of DNA replication, which is degraded during mitosis. Furthermore, the deregulation of AR activity is involved in the initiation of prostate cancer and contributes to castration resistant prostate cancer (CRPC). While AR degradation is known to occur primarily through a proteasome-mediated pathway, very little is known about how this process is regulated, especially in M phase. PC-1 is an androgen-responsive factor and expresses specificity in prostate cancer, with higher expression noted at G2/M. In this study, PC-1 was shown to interact with AR and E3 ligase CHIP (Carboxy-terminus of Hsc70 Interacting Protein) and to enhance AR/CHIP interactions, thereby decreasing AR stability. Moreover, PC-1 was found to act in conjunction with CHIP in the decreasing of AR via ubiquitination, with the subsequent degradation predominantly occurring during M phase. PC-1 was also found to repress AR transcriptional activity in androgen-dependent and androgen-independent prostate cancer cells and attenuate the growth inhibition of AR. In conclusion, these findings should provide new clues regarding the modulation of AR turnover and activity via PC-1 and reveals an essential role of PC-1 in AR signaling.
Collapse
Affiliation(s)
- Jian Wang
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Hui Zhang
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaoqing Zhang
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Peng Wang
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Hongtao Wang
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Fang Huang
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Chenyan Zhou
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jianguang Zhou
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Shanhu Li
- Laboratory of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| |
Collapse
|
25
|
Signaling regulation and role of filamin A cleavage in Ca2+-stimulated migration of androgen receptor-deficient prostate cancer cells. Oncotarget 2018; 8:3840-3853. [PMID: 27206800 PMCID: PMC5354799 DOI: 10.18632/oncotarget.9472] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/24/2016] [Indexed: 01/05/2023] Open
Abstract
Ca2+, a ubiquitous cellular signal, and filamin A, an actin-binding protein, play an important role in the regulation of cell adhesion, shape and motility. Using transwell filters to analyze cell migration, we found that extracellular Ca2+ (Cao2+) promotes the migration of androgen receptor (AR)-deficient and highly metastatic prostate cancer cell lines (DU145 and PC-3) compared to AR-positive and relatively less metastatic prostate cancer cells (LNCaP). Furthermore, we found that expression of filamin A is up-regulated in DU145 and PC-3 cells, and that Cao2+ significantly induces the cleavage of filamin A. Silencing expression of Ca2+-sensing receptor (CaR) and p115RhoGEF, and treating with leupeptin, a protease inhibitor, and ALLM, a calpain specific inhibitor, we further demonstrate that Cao2+-induced filamin A cleavage occurs via a CaR- p115RhoGEF-calpain dependent pathway. Our data show that Cao2+ via CaR- mediated signaling induces filamin A cleavage and promotes the migration in AR-deficient and highly metastatic prostate cancer cells.
Collapse
|
26
|
Mohammad OS, Nyquist MD, Schweizer MT, Balk SP, Corey E, Plymate S, Nelson PS, Mostaghel EA. Supraphysiologic Testosterone Therapy in the Treatment of Prostate Cancer: Models, Mechanisms and Questions. Cancers (Basel) 2017; 9:E166. [PMID: 29210989 PMCID: PMC5742814 DOI: 10.3390/cancers9120166] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Since Huggins defined the androgen-sensitive nature of prostate cancer (PCa), suppression of systemic testosterone (T) has remained the most effective initial therapy for advanced disease although progression inevitably occurs. From the inception of clinical efforts to suppress androgen receptor (AR) signaling by reducing AR ligands, it was also recognized that administration of T in men with castration-resistant prostate cancer (CRPC) could result in substantial clinical responses. Data from preclinical models have reproducibly shown biphasic responses to T administration, with proliferation at low androgen concentrations and growth inhibition at supraphysiological T concentrations. Many questions regarding the biphasic response of PCa to androgen treatment remain, primarily regarding the mechanisms driving these responses and how best to exploit the biphasic phenomenon clinically. Here we review the preclinical and clinical data on high dose androgen growth repression and discuss cellular pathways and mechanisms likely to be involved in mediating this response. Although meaningful clinical responses have now been observed in men with PCa treated with high dose T, not all men respond, leading to questions regarding which tumor characteristics promote response or resistance, and highlighting the need for studies designed to determine the molecular mechanism(s) driving these responses and identify predictive biomarkers.
Collapse
Affiliation(s)
- Osama S Mohammad
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- Faculty of Medicine, Benha University, Benha 13518, Egypt.
| | | | - Michael T Schweizer
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Stephen P Balk
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA.
| | - Stephen Plymate
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
27
|
Netchiporouk E, Gantchev J, Tsang M, Thibault P, Watters AK, Hughes JDM, Ghazawi FM, Woetmann A, Ødum N, Sasseville D, Litvinov IV. Analysis of CTCL cell lines reveals important differences between mycosis fungoides/Sézary syndrome vs. HTLV-1+ leukemic cell lines. Oncotarget 2017; 8:95981-95998. [PMID: 29221181 PMCID: PMC5707075 DOI: 10.18632/oncotarget.21619] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
HTLV-1 is estimated to affect ~20 million people worldwide and in ~5% of carriers it produces Adult T-Cell Leukemia/Lymphoma (ATLL), which can often masquerade and present with classic erythematous pruritic patches and plaques that are typically seen in Mycosis Fungoides (MF) and Sézary Syndrome (SS), the most recognized variants of Cutaneous T-Cell Lymphomas (CTCL). For many years the role of HTLV-1 in the pathogenesis of MF/SS has been hotly debated. In this study we analyzed CTCL vs. HTLV-1+ leukemic cells. We performed G-banding/spectral karyotyping, extensive gene expression analysis, TP53 sequencing in the 11 patient-derived HTLV-1+ (MJ and Hut102) vs. HTLV-1- (Myla, Mac2a, PB2B, HH, H9, Hut78, SZ4, Sez4 and SeAx) CTCL cell lines. We further tested drug sensitivities to commonly used CTCL therapies and studied the ability of these cells to produce subcutaneous xenograft tumors in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice. Our work demonstrates that unlike classic advanced MF/SS cells that acquire many ongoing balanced and unbalanced chromosomal translocations, HTLV-1+ CTCL leukemia cells are diploid and exhibit only a minimal number of non-specific chromosomal alterations. Our results indicate that HTLV-1 virus is likely not involved in the pathogenesis of classic MF/SS since it drives a very different pathway of lymphomagenesis based on our findings in these cells. This study also provides for the first time a comprehensive characterization of the CTCL cells with respect to gene expression profiling, TP53 mutation status, ability to produce tumors in mice and response to commonly used therapies.
Collapse
Affiliation(s)
| | - Jennifer Gantchev
- Division of Dermatology, McGill University, Montréal, Québec, Canada
| | - Matthew Tsang
- Division of Dermatology, University of Ottawa, Ottawa, Ontario, Canada
| | - Philippe Thibault
- Université de Sherbrooke Rnomics Platform, Sherbrooke, Québec, Canada
| | - Andrew K Watters
- Department of Pathology, McGill University Health Centre, Montreal, Québec, Canada
| | | | - Feras M Ghazawi
- Division of Dermatology, University of Ottawa, Ottawa, Ontario, Canada
| | - Anders Woetmann
- Department of International Health, Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Ødum
- Department of International Health, Immunology, and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Denis Sasseville
- Division of Dermatology, McGill University, Montréal, Québec, Canada
| | - Ivan V Litvinov
- Division of Dermatology, McGill University, Montréal, Québec, Canada.,Division of Dermatology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
28
|
McLean DT, Strand DW, Ricke WA. Prostate cancer xenografts and hormone induced prostate carcinogenesis. Differentiation 2017; 97:23-32. [PMID: 28923776 DOI: 10.1016/j.diff.2017.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/17/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022]
Abstract
Despite the advancement of transgenic and gene knockout animal models in the prostate cancer research, there is still a need for utilizing xenograft models. Xenografts can be grown in multiple sites/organs within immunocompromised animals such as mice and rats. Although prostate xenografts have been derived from many species, human cells and tissues are the most commonly used due to their potential clinical significance. Xenograft models that progress from one state or stage to another are commonly used to address important scientific questions including malignant transformation, metastatic spread, and castration resistance. Utilization of xenografts are commonly being used to assess the biology and genetics of prostate cancer, as well as, for therapeutic benefit. In addition to models for the study of prostate cancer, xenografts are also utilized as a tool in precision medicine where patient derived xenografts (PDX) can be grown in multiple animals and assessed for therapeutic efficacy. The popularity of such xenograft models and PDXs have led to availability of these resources through public and commercial institutions. In this review, we describe both traditional and emerging models of prostate cancer and their potential uses. Further development of current models and introduction of new models will likely provide new insights and better understanding of prostatic carcinogenesis and progression.
Collapse
Affiliation(s)
- Dalton T McLean
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Urology, University of Wisconsin-Madison, Madison, WI, USA; Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Institute, Dallas, TX, USA
| | - William A Ricke
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Urology, University of Wisconsin-Madison, Madison, WI, USA; George M. O'Brien Center of Research Excellence, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
29
|
Ahmad M, Sahabjada , Akhtar J, Hussain A, Badaruddeen , Arshad M, Mishra A. Development of a new rutin nanoemulsion and its application on prostate carcinoma PC3 cell line. EXCLI JOURNAL 2017; 16:810-823. [PMID: 28694767 PMCID: PMC5500835 DOI: 10.17179/excli2016-668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
Abstract
Biological effects of rutin bioactive are limited due to its poor oral bioavailability and its degradation in aqueous environments. For the purpose of bioenhancement, different nanoemulsion systems of rutin were developed by aqueous titration method using water as dispersion media. The nanoemulsion systems were characterized for surface morphology, droplet size, polydispersity index, zeta potential, in vitro release profile and the formulations were optimized. The anticancer potential of optimized nanoemulsion was evaluated by cells viability (MTT) assay, nuclear condensation, and ROS activity using human prostate cancer (PC3) cell line. On the basis of cell viability data the inhibitory concentration (IC50) value for optimized nanoemulsion formulation on PC3 cancer cells was found to be 11.8 μM. Fluorescent microscopic analysis and intracellular ROS generation demonstrated significant ROS induction that might lead to triggering the apoptosis pathway. In conclusion, developed nanoemulsion displayed significant efficacy against prostate carcinoma cells.
Collapse
Affiliation(s)
- Mohammad Ahmad
- Herbal Bioactive Research Laboratory, Faculty of Pharmacy, Integral University, Lucknow, India
| | - - Sahabjada
- Molecular Endocrinology Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| | - Juber Akhtar
- Herbal Bioactive Research Laboratory, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Arshad Hussain
- College of Pharmacy, King Khalid University, Abha, KSA. Formerly, Faculty of Pharmacy, Integral University, Lucknow, India
| | - - Badaruddeen
- Herbal Bioactive Research Laboratory, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Md Arshad
- Molecular Endocrinology Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| | - Anuradha Mishra
- Herbal Bioactive Research Laboratory, Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
30
|
Di Giacomo V, Tian TV, Mas A, Pecoraro M, Batlle-Morera L, Noya L, Martín-Caballero J, Ruberte J, Keyes WM. ΔNp63α promotes adhesion of metastatic prostate cancer cells to the bone through regulation of CD82. Oncogene 2017; 36:4381-4392. [PMID: 28368419 PMCID: PMC5543260 DOI: 10.1038/onc.2017.42] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 01/01/2017] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
ΔNp63α is a critical mediator of epithelial development and stem cell function in a variety of tissues including the skin and breast, while overexpression of ΔNp63α acts as an oncogene to drive tumor formation and cancer stem cell properties in squamous cell carcinoma. However, with regards to the prostate, while ΔNp63α is expressed in the basal stem cells of the mature gland, during adenocarcinoma development, its expression is lost and its absence is used to clinically diagnose the malignant state. Surprisingly, here we identify a sub-population of bone metastatic prostate cancer cells in the PC3 cell line that express ΔNp63α. Interestingly, we discovered that ΔNp63α favors adhesion and stem-like growth of these cells in the bone microenvironment. In addition, we show that these properties require expression of the target gene CD82. Together, this work uncovers a population of bone metastatic prostate cancer cells that express ΔNp63α, and provides important information about the mechanisms of bone metastatic colonization. Finally, we identify metastasis-promoting properties for the tetraspanin family member CD82.
Collapse
Affiliation(s)
- V Di Giacomo
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - T V Tian
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - A Mas
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - M Pecoraro
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Batlle-Morera
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Noya
- Department of Animal Health and Anatomy and Center for Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - J Ruberte
- Department of Animal Health and Anatomy and Center for Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - W M Keyes
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Development and Stem Cells program, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U964, Université de Strasbourg, Illkirch, France
| |
Collapse
|
31
|
Graham MK, Principessa L, Antony L, Meeker AK, Isaacs JT. Low p16 INK4a Expression in Early Passage Human Prostate Basal Epithelial Cells Enables Immortalization by Telomerase Expression Alone. Prostate 2017; 77:374-384. [PMID: 27859428 PMCID: PMC5548187 DOI: 10.1002/pros.23276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND There are two principal senescence barriers that must be overcome to successfully immortalize primary human epithelial cells in culture, stress-induced senescence, and replicative senescence. The p16INK4a /retinoblastoma protein (p16/Rb) pathway mediates stress-induced senescence, and is generally upregulated by primary epithelial cells in response to the artificial conditions from tissue culture. Replicative senescence is associated with telomere loss. Following each round of cell division, telomeres progressively shorten. Once telomeres shorten to a critical length, the DNA damage response pathway is activated, and the tumor suppressor p53 pathway triggers replicative senescence. Exogenous expression of telomerase in normal human epithelial cells extends the replicative capacity of cells, and in some cases, immortalizes cells. However reliable immortalization of epithelial cells usually requires telomerase activity coupled with inactivation of the p16/Rb pathway. METHODS A lentiviral vector, pLOX-TERT-iresTK (Addgene #12245), containing a CMV promoter upstream of a bicistronic coding cassette that includes loxP sites flanking the catalytic subunit of human telomerase gene (TERT) and herpes simplex virus type-1 thymidine kinase gene (HSV1-tk) was used to transduce normal prostate basal epithelial cells (PrECs) initiated in cell culture from prostate cancer patients undergoing radical prostatectomies. RESULTS Transduction of early (i.e., <7) passage PrECs with TERT led to successful immortalization. However, attempts to immortalize late (i.e., >7) passage PrECs were unsuccessful. Late passage PrECs, which acquired elevated p16, were unable to overcome the senescence barrier. Immortalized PrECs (TERT-PrECs) retained a normal male karyotype and low p16 expression. Additionally, TERT-PrECs were non-tumorigenic when inoculated into intact male immunodeficient NSG mice. CONCLUSIONS The present studies document that early passage human PrECs have sufficiently low p16 to permit immortalization by TERT expression alone. TERT-PrECs developed using this transduction approach provides an appropriate and experimentally facile model for clarifying the molecular mechanism(s) involved in both immortalization of human PrECs, as well as identifying genetic/epigenetic "drivers" for conversion of these immortalized non-tumorigenic cells into fully lethal prostate cancers. Notably, loxP sites flank the exogenous TERT gene in the TERT-PrECs. Cre recombinase can be used to excise TERT, and resolve whether TERT expression is required for these cells to be fully transformed into lethal cancer. Prostate 77: 374-384, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mindy Kim Graham
- Department of Pathology, John Hopkins University School of Medicine, Baltimore, Maryland
| | - Lorenzo Principessa
- Chemical Therapeutic Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Lizamma Antony
- Chemical Therapeutic Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Alan K. Meeker
- Departments of Pathology, Oncology and Urology, John Hopkins University School of Medicine, Baltimore, Maryland
| | - John T. Isaacs
- Chemical Therapeutic Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Correspondence to: Dr. John T. Isaacs, Chemical Therapeutic Program, Bunting-Blaustein CRB1, 1650 Orleans Street, Baltimore, MD 21231.
| |
Collapse
|
32
|
Palethorpe HM, Drew PA, Smith E. Androgen Signaling in Esophageal Adenocarcinoma Cell Lines In Vitro. Dig Dis Sci 2017; 62:3402-3414. [PMID: 29052817 PMCID: PMC5694516 DOI: 10.1007/s10620-017-4794-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/06/2017] [Indexed: 01/11/2023]
Abstract
BACKGROUND We showed previously that nuclear localization of the androgen receptor (AR) and expression of the androgen-responsive gene FK506-binding protein 5 (FKBP5) in esophageal adenocarcinoma (EAC) tissues were associated with decreased patient survival, suggesting a role for androgens in this cancer. AIM To investigate the effect of the AR ligand 5α-dihydrotestosterone (DHT) on AR-expressing EAC cell lines in vitro. METHODS AND RESULTS In tissue resection specimens from EAC patients, FKBP5 expression was positively associated with proliferation as measured by Ki-67 expression. We stably transduced AR into three AR-negative EAC cell lines, OE33, JH-EsoAd1, and OE19, to investigate androgen signaling in vitro. In the AR-expressing cell lines, 10 nM DHT, the concentration typically used to study AR signaling, induced changes in the expression of androgen-responsive genes and inhibited proliferation by inducing cell cycle arrest and senescence. At lower DHT concentrations near the half maximal inhibitory concentration (IC50), the AR-expressing cell lines proliferated and there were changes in the expression of androgen-responsive genes. In direct co-culture with cancer-associated fibroblast-like PShTert myofibroblasts, 10 nM DHT induced changes in the expression of androgen-responsive genes but did not inhibit proliferation. CONCLUSIONS This is the first study to show that EAC cell lines respond to androgen in vitro. Proliferation together with the expression of androgen-responsive genes was dependent on the concentration of DHT, or the presence of a permissive microenvironment, consistent with observations in the tissues. These findings are consistent with a role for androgen signaling in EAC.
Collapse
Affiliation(s)
- Helen M. Palethorpe
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia
| | - Paul A. Drew
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia ,0000 0004 0367 2697grid.1014.4School of Nursing and Midwifery, Flinders University, PO Box 2100, Adelaide, SA 5001 Australia
| | - Eric Smith
- 0000 0004 1936 7304grid.1010.0Solid Cancer Regulation Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, The University of Adelaide, 28 Woodville Rd, Woodville South, SA 5011 Australia ,0000 0004 0486 659Xgrid.278859.9Department of Medical Oncology, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville South, SA 5011 Australia
| |
Collapse
|
33
|
Komaragiri SK, Bostanthirige DH, Morton DJ, Patel D, Joshi J, Upadhyay S, Chaudhary J. ID4 promotes AR expression and blocks tumorigenicity of PC3 prostate cancer cells. Biochem Biophys Res Commun 2016; 478:60-66. [PMID: 27462022 PMCID: PMC4991035 DOI: 10.1016/j.bbrc.2016.07.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/21/2016] [Indexed: 01/23/2023]
Abstract
Deregulation of tumor suppressor genes is associated with tumorigenesis and the development of cancer. In prostate cancer, ID4 is epigenetically silenced and acts as a tumor suppressor. In normal prostate epithelial cells, ID4 collaborates with androgen receptor (AR) and p53 to exert its tumor suppressor activity. Previous studies have shown that ID4 promotes tumor suppressive function of AR whereas loss of ID4 results in tumor promoter activity of AR. Previous study from our lab showed that ectopic ID4 expression in DU145 attenuates proliferation and promotes AR expression suggesting that ID4 dependent AR activity is tumor suppressive. In this study, we examined the effect of ectopic expression of ID4 on highly malignant prostate cancer cell, PC3. Here we show that stable overexpression of ID4 in PC3 cells leads to increased apoptosis and decreased cell proliferation and migration. In addition, in vivo studies showed a decrease in tumor size and volume of ID4 overexpressing PC3 cells, in nude mice. At the molecular level, these changes were associated with increased androgen receptor (AR), p21, and AR dependent FKBP51 expression. At the mechanistic level, ID4 may regulate the expression or function of AR through specific but yet unknown AR co-regulators that may determine the final outcome of AR function.
Collapse
Affiliation(s)
- Shravan Kumar Komaragiri
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Dhanushka H Bostanthirige
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Derrick J Morton
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Divya Patel
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Jugal Joshi
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Sunil Upadhyay
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States
| | - Jaideep Chaudhary
- Department of Biology and Center for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, GA 30314, United States.
| |
Collapse
|
34
|
Ebron JS, Shukla GC. Molecular characterization of a novel androgen receptor transgene responsive to MicroRNA mediated post-transcriptional control exerted via 3'-untranslated region. Prostate 2016; 76:834-44. [PMID: 26988939 DOI: 10.1002/pros.23174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/16/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Androgen Receptor (AR) gene is associated with Prostate cancer (PCa) and hence targeting androgen-and AR-signaling axis remains the most promising primary therapeutic option to treat the disease. The AR mRNA has a 6.8 kb long 3'-untranslated region (UTR) which harbors several experimentally validated and numerous predicted miRNA binding sites. AR 3'-UTR is likely to positively or negatively regulate AR expression by interacting with miRNAs and possibly other trans-acting auxiliary factors including 3'-UTR RNA binding proteins. In this context, systematic understanding of the regulatory role of AR 3'-UTR in intrinsic post-transcriptional control of AR gene expression is of significance to understand AR related diseases including PCa. METHODS In this study, we have constructed a heterologous reporter system in which Firefly luciferase and AR expression is experimentally influenced by the presence of AR 3'-UTR and its interactions with ectopically expressing miRNA. RESULTS The expression of AR 3'-UTR containing reporters, including the Firefly luciferase and the AR open reading frame (ORF) were repressed by the overexpression of miR-488* mimics. In addition, the AR expressed from 3'-UTR containing expression vectors was fully functional in its transactivation function as determined by a prostate specific antigen (PSA) reporter assay. Further, by using confocal microscopy we also demonstrate that AR can translocate to the nucleus upon DHT activation confirming the functional ability of AR. CONCLUSIONS AR transgenes with AR 3'-UTR fragments closely resemble the endogenous AR expression than any other previously characterized AR expression constructs. The 3'-UTR containing AR expression system is amiable to post-transcriptional manipulations including miRNA mediated repression of AR expression. This AR reporter system has the potential to be used in determining specificity of AR targeting miRNAs and their role in AR functional regulatory networks. Prostate 76:834-844, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jey Sabith Ebron
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
- Department of Biological Sciences, Cleveland State University, Cleveland, Ohio
| | - Girish C Shukla
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
- Department of Biological Sciences, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
35
|
Chen C, Dienhart JA, Bolton EC. Androgen-Sensitized Apoptosis of HPr-1AR Human Prostate Epithelial Cells. PLoS One 2016; 11:e0156145. [PMID: 27203692 PMCID: PMC4874596 DOI: 10.1371/journal.pone.0156145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/10/2016] [Indexed: 01/27/2023] Open
Abstract
Androgen receptor (AR) signaling is crucial to the development and homeostasis of the prostate gland, and its dysregulation mediates common prostate pathologies. The mechanisms whereby AR regulates growth suppression and differentiation of luminal epithelial cells in the prostate gland and proliferation of malignant versions of these cells have been investigated in human and rodent adult prostate. However, the cellular stress response of human prostate epithelial cells is not well understood, though it is central to prostate health and pathology. Here, we report that androgen sensitizes HPr-1AR and RWPE-AR human prostate epithelial cells to cell stress agents and apoptotic cell death. Although 5α-dihydrotestosterone (DHT) treatment alone did not induce cell death, co-treatment of HPr-1AR cells with DHT and an apoptosis inducer, such as staurosporine (STS), TNFt, or hydrogen peroxide, synergistically increased cell death in comparison to treatment with each apoptosis inducer by itself. We found that the synergy between DHT and apoptosis inducer led to activation of the intrinsic/mitochondrial apoptotic pathway, which is supported by robust cleavage activation of caspase-9 and caspase-3. Further, the dramatic depolarization of the mitochondrial membrane potential that we observed upon co-treatment with DHT and STS is consistent with increased mitochondrial outer membrane permeabilization (MOMP) in the pro-apoptotic mechanism. Interestingly, the synergy between DHT and apoptosis inducer was abolished by AR antagonists and inhibitors of transcription and protein synthesis, suggesting that AR mediates pro-apoptotic synergy through transcriptional regulation of MOMP genes. Expression analysis revealed that pro-apoptotic genes (BCL2L11/BIM and AIFM2) were DHT-induced, whereas pro-survival genes (BCL2L1/BCL-XL and MCL1) were DHT-repressed. Hence, we propose that the net effect of these AR-mediated expression changes shifts the balance of BCL2-family proteins, such that androgen signaling sensitizes mitochondria to apoptotic signaling, thus rendering HPr-1AR more vulnerable to cell death signals. Our study offers insight into AR-mediated regulation of prostate epithelial cell death signaling.
Collapse
Affiliation(s)
- Congcong Chen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jason A. Dienhart
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Eric C. Bolton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
36
|
Asuthkar S, Velpula KK, Elustondo PA, Demirkhanyan L, Zakharian E. TRPM8 channel as a novel molecular target in androgen-regulated prostate cancer cells. Oncotarget 2016; 6:17221-36. [PMID: 25980497 PMCID: PMC4627303 DOI: 10.18632/oncotarget.3948] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022] Open
Abstract
The cold and menthol receptor TRPM8 is highly expressed in prostate and prostate cancer (PC). Recently, we identified that TRPM8 is as an ionotropic testosterone receptor. The TRPM8 mRNA is expressed in early prostate tumors with high androgen levels, while anti-androgen therapy greatly reduces its expression. Here, from the chromatin-immunoprecipitation (ChIP) analysis, we found that an androgen response element (ARE) mediates androgen regulation of trpm8. Furthermore, using immunofluorescence, calcium-imaging and planar lipid bilayers, we identified that TRPM8 channel is functionally regulated by androgens in the prostate. Although TRPM8 mRNA is expressed at high levels, we found that the TRPM8 protein undergoes ubiquitination and degradation in PC cells. The mass-spectrometry analysis of TRPM8, immunoprecipitated from LNCaP cells identified ubiquitin-like modifier-activating enzyme 1 (UBA1). PYR-41, a potent inhibitor of initial enzyme in the ubiquitination cascade, UBA1, increased TRPM8 activity on the plasma membrane (PM) of LNCaP cells. Furthermore, PYR-41-mediated PMTRPM8 activity was accompanied by enhanced activation of p53 and Caspase-9. Interestingly, we found that the trpm8 promoter possesses putative binding sites for p53 and that the overexpression of p53 increased the TRPM8 mRNA levels. In addition to the genomic regulation of TRPM8 by AR and p53, our findings indicate that the testosterone-induced PMTRPM8 activity elicits Ca2+ uptake, subsequently causing apoptotic cell death. These findings support the strategy of rescuing PMTRPM8 expression as a new therapeutic application through the regulation of PC cell growth and proliferation.
Collapse
Affiliation(s)
- Swapna Asuthkar
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | - Kiran Kumar Velpula
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | | | - Lusine Demirkhanyan
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| | - Eleonora Zakharian
- University of Illinois College of Medicine, Department of Cancer Biology and Pharmacology, Peoria, IL, USA
| |
Collapse
|
37
|
Kao CH, Bishop KS, Xu Y, Han DY, Murray PM, Marlow GJ, Ferguson LR. Identification of Potential Anticancer Activities of Novel Ganoderma lucidum Extracts Using Gene Expression and Pathway Network Analysis. GENOMICS INSIGHTS 2016; 9:1-16. [PMID: 27006591 PMCID: PMC4778854 DOI: 10.4137/gei.s32477] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/22/2015] [Accepted: 10/24/2015] [Indexed: 01/07/2023]
Abstract
Ganoderma lucidum (lingzhi) has been used for the general promotion of health in Asia for many centuries. The common method of consumption is to boil lingzhi in water and then drink the liquid. In this study, we examined the potential anticancer activities of G. lucidum submerged in two commonly consumed forms of alcohol in East Asia: malt whiskey and rice wine. The anticancer effect of G. lucidum, using whiskey and rice wine-based extraction methods, has not been previously reported. The growth inhibition of G. lucidum whiskey and rice wine extracts on the prostate cancer cell lines, PC3 and DU145, was determined. Using Affymetrix gene expression assays, several biologically active pathways associated with the anticancer activities of G. lucidum extracts were identified. Using gene expression analysis (real-time polymerase chain reaction [RT-PCR]) and protein analysis (Western blotting), we confirmed the expression of key genes and their associated proteins that were initially identified with Affymetrix gene expression analysis.
Collapse
Affiliation(s)
- Chi H.J. Kao
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Karen S. Bishop
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Yuanye Xu
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Dug Yeo Han
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Pamela M. Murray
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Gareth J. Marlow
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Lynnette R. Ferguson
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
38
|
Su Q, Xin L. Notch signaling in prostate cancer: refining a therapeutic opportunity. Histol Histopathol 2016; 31:149-57. [PMID: 26521657 PMCID: PMC4822406 DOI: 10.14670/hh-11-685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Notch is an evolutionarily conserved signaling pathway that plays a critical role in specifying cell fate and regulating tissue homeostasis and carcinogenesis. Studies using organ cultures and genetically engineered mouse models have demonstrated that Notch signaling regulates prostate development and homeostasis. However, the role of the Notch signaling pathway in prostate cancer remains inconclusive. Many published studies have documented consistent deregulation of major Notch signaling components in human prostate cancer cell lines, mouse models for prostate cancers, and human prostate cancer specimens at both the mRNA and the protein levels. However, functional studies in human cancer cells by modulation of Notch pathway elements suggest both tumor suppressive and oncogenic roles of Notch. These controversies may originate from our inadequate understanding of the regulation of Notch signaling under versatile genetic contexts, and reflect the multifaceted and pleiotropic roles of Notch in regulating different aspects of prostate cancer cell biology, such as proliferation, metastasis, and chemo-resistance. Future comprehensive studies using various mouse models for prostate cancer may help clarify the role of Notch signaling in prostate cancer and provide a solid basis for determining whether and how Notch should be employed as a therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Qingtai Su
- Department of Molecular and Cellular Biology, Baylor College of Medicine, and Graduate Program in Integrative Molecular and Biomedical Sciences, Houston, Texas, USA
| | - Li Xin
- Department of Molecular and Cellular Biology, Department of Pathology and Immunology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
39
|
Deng Q, Tang DG. Androgen receptor and prostate cancer stem cells: biological mechanisms and clinical implications. Endocr Relat Cancer 2015; 22:T209-20. [PMID: 26285606 PMCID: PMC4646167 DOI: 10.1530/erc-15-0217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2015] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) contains phenotypically and functionally distinct cells, and this cellular heterogeneity poses clinical challenges as the distinct cell types likely respond differently to various therapies. Clonal evolution, driven by genetic instability, and intraclonal cancer cell diversification, driven by cancer stem cells (CSCs), together create tumor cell heterogeneity. In this review, we first discuss PCa stem cells (PCSCs) and heterogeneity of androgen receptor (AR) expression in primary, metastatic, and treatment-failed PCa. Based on literature reports and our own studies, we hypothesize that, whereas PCSCs in primary and untreated tumors and models are mainly AR(-), PCSCs in CRPCs could be either AR(+) or AR(-/lo). We illustrate the potential mechanisms AR(+) and AR(-) PCSCs may employ to propagate PCa at the population level, mediate therapy resistance, and metastasize. As a result, targeting AR alone may not achieve long-lasting therapeutic efficacy. Elucidating the roles of AR and PCSCs should provide important clues to designing novel personalized combinatorial therapeutic protocols targeting both AR(+) and AR(-) PCa cells.
Collapse
Affiliation(s)
- Qu Deng
- Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Dean G Tang
- Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
40
|
Sorrenti V, Vanella L, Acquaviva R, Cardile V, Giofrè S, Di Giacomo C. Cyanidin induces apoptosis and differentiation in prostate cancer cells. Int J Oncol 2015; 47:1303-1310. [PMID: 26315029 DOI: 10.3892/ijo.2015.3130] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/11/2015] [Indexed: 11/05/2022] Open
Abstract
Several natural antioxidants, including anthocyanins, have been reported to have chemotherapeutic activity in vivo and in vitro. The aim of the present study was to delineate the anti-proliferative activity and the cytodifferentiation properties mediated by cyanidin-3-O-β-glucopyranoside (C3G) treatment in the DU145 and LnCap human prostatic cancer cell lines. C3G produced anti-proliferative effects through activation of caspase-3 and induction of p21 protein expression. The reduced cell viability was associated with a clear increase of DNA fragmentation in both cell lines after C3G treatment. Since LnCap and DU145 exhibited differences in sensitivity to C3G treatment, the redox state of these cells was further investigated by estimating the levels of ROS and GSH. C3G antioxidant activity was confirmed only in DU145 cell line. Treatment with C3G increased the levels of tumor suppressor P75NGFR, indicating a possible role of C3G in the acquisition of a normal-like cell phenotype. Results reported in the present study demonstrate that C3G, the most abundant anthocyanin in diet, may represent a new approach and highly effective strategy in reducing carcinogenesis. C3G may be considered a new therapeutic agent with both anti-proliferative and pro-differentiation properties.
Collapse
Affiliation(s)
- Valeria Sorrenti
- Department of Drug Science, Biochemistry Section, University of Catania, I-95125 Catania, Italy
| | - Luca Vanella
- Department of Drug Science, Biochemistry Section, University of Catania, I-95125 Catania, Italy
| | - Rosaria Acquaviva
- Department of Drug Science, Biochemistry Section, University of Catania, I-95125 Catania, Italy
| | - Venera Cardile
- Department of Bio-Medical Sciences, Physiology Section, University of Catania, I-95125 Catania, Italy
| | - Salvatore Giofrè
- Department of 'Scienze del Farmaco e Prodotti per la Salute', University of Messina, I-98168 Messina, Italy
| | - Claudia Di Giacomo
- Department of Drug Science, Biochemistry Section, University of Catania, I-95125 Catania, Italy
| |
Collapse
|
41
|
Kim YC, Chen C, Bolton EC. Androgen Receptor-Mediated Growth Suppression of HPr-1AR and PC3-Lenti-AR Prostate Epithelial Cells. PLoS One 2015; 10:e0138286. [PMID: 26372468 PMCID: PMC4570807 DOI: 10.1371/journal.pone.0138286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/29/2015] [Indexed: 12/19/2022] Open
Abstract
The androgen receptor (AR) mediates the developmental, physiologic, and pathologic effects of androgens including 5α-dihydrotestosterone (DHT). However, the mechanisms whereby AR regulates growth suppression and differentiation of luminal epithelial cells in the prostate gland and proliferation of malignant versions of these cells are not well understood, though they are central to prostate development, homeostasis, and neoplasia. Here, we identify androgen-responsive genes that restrain cell cycle progression and proliferation of human prostate epithelial cell lines (HPr-1AR and PC3-Lenti-AR), and we investigate the mechanisms through which AR regulates their expression. DHT inhibited proliferation of HPr-1AR and PC3-Lenti-AR, and cell cycle analysis revealed a prolonged G1 interval. In the cell cycle, the G1/S-phase transition is initiated by the activity of cyclin D and cyclin-dependent kinase (CDK) complexes, which relieve growth suppression. In HPr-1AR, cyclin D1/2 and CDK4/6 mRNAs were androgen-repressed, whereas CDK inhibitor, CDKN1A, mRNA was androgen-induced. The regulation of these transcripts was AR-dependent, and involved multiple mechanisms. Similar AR-mediated down-regulation of CDK4/6 mRNAs and up-regulation of CDKN1A mRNA occurred in PC3-Lenti-AR. Further, CDK4/6 overexpression suppressed DHT-inhibited cell cycle progression and proliferation of HPr-1AR and PC3-Lenti-AR, whereas CDKN1A overexpression induced cell cycle arrest. We therefore propose that AR-mediated growth suppression of HPr-1AR involves cyclin D1 mRNA decay, transcriptional repression of cyclin D2 and CDK4/6, and transcriptional activation of CDKN1A, which serve to decrease CDK4/6 activity. AR-mediated inhibition of PC3-Lenti-AR proliferation occurs through a similar mechanism, albeit without down-regulation of cyclin D. Our findings provide insight into AR-mediated regulation of prostate epithelial cell proliferation.
Collapse
Affiliation(s)
- Young-Chae Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Congcong Chen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Eric C. Bolton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
42
|
Mitani T, Minami M, Harada N, Ashida H, Yamaji R. Autophagic degradation of the androgen receptor mediated by increased phosphorylation of p62 suppresses apoptosis in hypoxia. Cell Signal 2015; 27:1994-2001. [PMID: 26186973 DOI: 10.1016/j.cellsig.2015.07.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/25/2022]
Abstract
Prostate cancer grows under hypoxic conditions. Hypoxia decreases androgen receptor (AR) protein levels. However, the molecular mechanism remains unclear. Here, we report that p62-mediated autophagy degrades AR protein and suppresses apoptosis in prostate cancer LNCaP cells in hypoxia. In LNCaP cells, hypoxia decreased AR at the protein level, but not at the mRNA level. Hypoxia-induced AR degradation was inhibited not only by knockdown of LC3, a key component of the autophagy machinery, but also by knockdown of p62. Depletion of p62 enhanced hypoxia-induced poly(ADP-ribose) polymerase cleavage and caspase-3 cleavage, markers of apoptosis, whereas simultaneous knockdown of p62 and AR suppressed hypoxia-induced apoptosis. Hypoxia increased the formation of a cytosolic p62-AR complex and enhanced sequestration of AR from the nucleus. Formation of this complex was promoted by the increased phosphorylation of serine 403 in the ubiquitin-associated domain of p62 during hypoxia. An antioxidant and an AMP-activated protein kinase (AMPK) inhibitor reduced hypoxia-induced p62 phosphorylation at serine 403 and suppressed hypoxia-induced complex formation between AR and p62. These results demonstrate that hypoxia enhances the complex formation between p62 and AR by promoting phosphorylation of p62 at serine 403, probably through activating AMPK, and that p62-mediated autophagy degrades AR protein for cell survival in hypoxia.
Collapse
Affiliation(s)
- Takakazu Mitani
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 5998531, Japan; Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 6578501, Japan
| | - Masato Minami
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 5998531, Japan
| | - Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 5998531, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 6578501, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 5998531, Japan.
| |
Collapse
|
43
|
Cardoso HJ, Vaz CV, Correia S, Figueira MI, Marques R, Maia CJ, Socorro S. Paradoxical and contradictory effects of imatinib in two cell line models of hormone-refractory prostate cancer. Prostate 2015; 75:923-935. [PMID: 25786656 DOI: 10.1002/pros.22976] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/13/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND Imatinib mesylate is a chemotherapeutic drug that inhibits the tyrosine kinase activity of c-KIT and has been successfully used to treat leukemias and some solid tumors. However, its application for treatment of hormone-refractory prostate cancer (HRPC) has shown modest effectiveness and did not follow the outcomes in cultured cells or animal models. Moreover, the molecular pathways by which imatinib induces cytotoxicity in prostate cancer cells are poorly characterized. METHODS Two cell line models of HRPC (DU145 and PC3) were exposed to 20 μM of imatinib for 6-72 hr. MTS assay was used to assess cell viability during the course of experiment. Gene expression analysis of c-KIT, cell-cycle and apoptosis regulators, and angiogenic factors was determined by means of real-time PCR, western blot, and/or immunocytochemistry. The enzymatic activity of the apoptosis effector, caspase-3, was determined by a colorimetric assay. RESULTS Imatinib significantly decreased the viability of DU145 cells but paradoxically augmented the viability of PC3 cells. DU145 cells displayed diminished expression of anti-apoptotic Bcl-2 protein and augmented levels of caspase-8 and -9, as well as, increased enzymatic activity of caspase-3 in response to imatinib. No differences existed on the expression levels of apoptosis-related proteins in PC3 cells treated with imatinib, though the activity of caspase-3 was decreased. The mRNA levels of angiogenic factor VEGF were decreased in DU145-treated cells, whereas an opposite effect was seen in PC3. In addition, it was shown that DU145 and PC3 cells present a differential expression of c-KIT protein variants. CONCLUSION DU145 and PC3 cells displayed a contradictory behavior in response to imatinib, which was underpinned by a distinct expression pattern (or activity) of target regulators of cell-cycle, apoptosis, and angiogenesis. The paradoxical effect of imatinib in PC3 cells may be related with the differential expression of c-KIT protein variants. Moreover, the present findings helped to understand the discrepancies in the efficacy of imatinib as therapeutic option in HRPC.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI, Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | | | | | |
Collapse
|
44
|
Ellis L, Ku SY, Ramakrishnan S, Lasorsa E, Azabdaftari G, Godoy A, Pili R. Combinatorial antitumor effect of HDAC and the PI3K-Akt-mTOR pathway inhibition in a Pten defecient model of prostate cancer. Oncotarget 2014; 4:2225-36. [PMID: 24163230 PMCID: PMC3926822 DOI: 10.18632/oncotarget.1314] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increased expression of histone deacetylases (HDACs) and activation of the PI3K-Akt-mTORC1 pathway are common aberrations in prostate cancer (PCa). For this reason, inhibition of such targets is an exciting avenue for the development of novel therapeutic strategies to treat patients with advanced PCa. Previous reports demonstrated that HDAC inhibitors (HDACi) increases DNA damage and induce greater apoptosis in PCa cell lines that express androgen receptor (AR). In this study we utilized the AR negative PCa cell line and observed that re-expression of AR (PC3-AR) results in greater levels of apoptosis when treated with the pan-DACi, panobinostat (PAN). PAN mediated apoptosis in PC3 and PC3-AR cells was associated with increased levels of double strand DNA breaks, indicated by p-ɣH2AX. Further, PAN treatment in PC3-AR cells resulted in moderate attenuation of the ATM-Akt-ERK DNA damage response pathway. For this reason, we combined PAN with the dual PI3K-mTOR inhibitor, BEZ235. Combination of PAN with BEZ235 resulted in significant attenuation of the DNA damage repair protein ATM and significantly increased anti-tumor activity compared to each single treatment. Overall, superior anti-tumor activity with combination of PAN with BEZ235 was independent of AR status. These findings suggest that this therapeutic strategy should be further developed in clinical trials.
Collapse
Affiliation(s)
- Leigh Ellis
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo NY
| | | | | | | | | | | | | |
Collapse
|
45
|
Ta HQ, Gioeli D. The convergence of DNA damage checkpoint pathways and androgen receptor signaling in prostate cancer. Endocr Relat Cancer 2014; 21:R395-407. [PMID: 25096064 PMCID: PMC4382101 DOI: 10.1530/erc-14-0217] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is increasingly clear that castration-resistant prostate cancer (PCa) is dependent on the androgen receptor (AR). This has led to the use of anti-androgen therapies that reduce endogenous steroid hormone production as well as the use of AR antagonists. However, the AR does not act in isolation and integrates with a milieu of cell-signaling proteins to affect cell biology. It is well established that cancer is a genetic disease resulting from the accumulation of mutations and chromosomal translocations that enables cancer cells to survive, proliferate, and disseminate. To maintain genomic integrity, there exists conserved checkpoint signaling pathways to facilitate cell cycle delay, DNA repair, and/or apoptosis in response to DNA damage. The AR interacts with, affects, and is affected by these DNA damage-response proteins. This review will focus on the connections between checkpoint signaling and the AR in PCa. We will describe what is known about how components of checkpoint signaling regulate AR activity and what questions still face the field.
Collapse
Affiliation(s)
- Huy Q Ta
- Department of MicrobiologyImmunology, and Cancer BiologyUVA Cancer CenterUniversity of Virginia, PO Box 800734, Charlottesville, Virginia 22908, USA
| | - Daniel Gioeli
- Department of MicrobiologyImmunology, and Cancer BiologyUVA Cancer CenterUniversity of Virginia, PO Box 800734, Charlottesville, Virginia 22908, USA Department of MicrobiologyImmunology, and Cancer BiologyUVA Cancer CenterUniversity of Virginia, PO Box 800734, Charlottesville, Virginia 22908, USA
| |
Collapse
|
46
|
Antony L, van der Schoor F, Dalrymple SL, Isaacs JT. Androgen receptor (AR) suppresses normal human prostate epithelial cell proliferation via AR/β-catenin/TCF-4 complex inhibition of c-MYC transcription. Prostate 2014; 74:1118-31. [PMID: 24913829 PMCID: PMC4856018 DOI: 10.1002/pros.22828] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 05/02/2014] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Physiologic testosterone continuously stimulates prostate stromal cell secretion of paracrine growth factors (PGFs), which if unopposed would induce hyperplastic overgrowth of normal prostate epithelial cells (PrECs). METHODS Lentiviral shRNA stable knock down of c-MYC, β-catenin, or TCF-4 completely inhibits normal (i.e., non-transformed) human PrECs growth. c-MYC enhancer driven reporter expression and growth is inhibited by two chemically distinct molecules, which prevent β-catenin signaling either by blocking TCF-4 binding (i.e., toxoflavin) or by stimulating degradation (i.e., AVX939). Recombinant DKK1 protein at a dose, which inhibits activation of canonical Wnt signaling does not inhibit PrEC growth. Nuclear β-catenin translocation and PrEC growth is prevented by both lack of PGFs or Akt inhibitor-I. Growth inhibition induced by lack of PGFs, toxoflavin, or Akt inhibitor-I is overcome by constitutive c-MYC transcription. RESULTS In the presence of continuous PGF signaling, PrEC hyperplasia is prevented by androgen binding to AR suppressing c-MYC transcription, resulting in G0 arrest/terminal differentiation independent of Rb, p21, p27, FoxP3, or down regulation of growth factors receptors and instead involves androgen-induced formation of AR/β-catenin/TCF-4 complexes, which suppress c-MYC transcription. Such suppression does not occur when AR is mutated in its zinc-finger binding domain. DISCUSSION Proliferation of non-transformed human PrECs is dependent upon c-MYC transcription via formation/binding of β-catenin/TCF-4 complexes at both 5' and 3' c-MYC enhancers stimulated by Wnt-independent, PGF induced Akt signaling. In the presence of continuous PGF signaling, PrEC hyperplasia is prevented by androgen-induced formation of AR/β-catenin/TCF-4 complexes, which retains binding to 3' c-MYC enhancer, but now suppresses c-MYC transcription.
Collapse
Affiliation(s)
- Lizamma Antony
- Chemical Therapeutics Program and Prostate Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Urology, The Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Correspondence to: Lizamma Antony, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans St., Baltimore, MD 21287.
| | - Freek van der Schoor
- Chemical Therapeutics Program and Prostate Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Urology, The Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Susan L. Dalrymple
- Chemical Therapeutics Program and Prostate Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Urology, The Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John T. Isaacs
- Chemical Therapeutics Program and Prostate Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Urology, The Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
Vander Griend DJ, Litvinov IV, Isaacs JT. Conversion of androgen receptor signaling from a growth suppressor in normal prostate epithelial cells to an oncogene in prostate cancer cells involves a gain of function in c-Myc regulation. Int J Biol Sci 2014; 10:627-42. [PMID: 24948876 PMCID: PMC4062956 DOI: 10.7150/ijbs.8756] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/12/2014] [Indexed: 12/22/2022] Open
Abstract
In normal prostate, androgen-dependent androgen receptor (AR) signaling within prostate stromal cells induces their secretion of paracrine factors, termed “andromedins” which stimulate growth of the epithelial cells. The present studies demonstrate that androgen-dependent andromedin-driven growth stimulation is counter-balanced by androgen-induced AR signaling within normal adult prostate epithelial cells resulting in terminal G0 growth arrest coupled with terminal differentiation into ΔNp63-negative, PSA-expressing secretory luminal cells. This cell autonomous AR-driven terminal differentiation requires DNA-binding of the AR protein, is associated with decreases in c-Myc m-RNA and protein, are coupled with increases in p21, p27, and SKP-2 protein expression, and does not require functional p53. These changes result in down-regulation of Cyclin D1 protein and RB phosphoryation. shRNA knockdown documents that neither RB, p21, p27 alone or in combination are required for such AR-induced G0 growth arrest. Transgenic expression of a constitutive vector to prevent c-Myc down-regulation overrides AR-mediated growth arrest in normal prostate epithelial cells, which documents that AR-induced c-Myc down-regulation is critical in terminal growth arrest of normal prostate epithelial cells. In contrast, in prostate cancer cells, androgen-induced AR signaling paradoxically up-regulates c-Myc expression and stimulates growth as documented by inhibition of both of these responses following exposure to the AR antagonist, bicalutamide. These data document that AR signaling is converted from a growth suppressor in normal prostate epithelial cells to an oncogene in prostate cancer cells during prostatic carcinogenesis and that this conversion involves a gain of function for regulation of c-Myc expression.
Collapse
Affiliation(s)
- Donald J Vander Griend
- 1. Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. ; 3. The Brady Urological Institute, Johns Hopkins
| | - Ivan V Litvinov
- 1. Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. ; 2. Cellular and Molecular Medicine Graduate Program at Johns Hopkins
| | - John T Isaacs
- 1. Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. ; 2. Cellular and Molecular Medicine Graduate Program at Johns Hopkins. ; 3. The Brady Urological Institute, Johns Hopkins
| |
Collapse
|
48
|
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: 7.5] [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
|
49
|
Liu PY, Lin SZ, Sheu JJC, Lin CT, Lin PC, Chou YW, Huang MH, Chiou TW, Harn HJ. Regulation of androgen receptor expression by Z-isochaihulactone mediated by the JNK signaling pathway and might be related to cytotoxicity in prostate cancer. Prostate 2013; 73:531-41. [PMID: 23038474 DOI: 10.1002/pros.22593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 08/30/2012] [Indexed: 11/11/2022]
Abstract
BACKGROUND The androgen receptor (AR) is a main therapeutic target for treatment of prostate cancer (PCa). The natural compound isochaihulactone (K8), which has a chiral center ring and two racemic forms (E-K8 and Z-K8), has anti-tumor effects on multiple cancer types both in vitro and in vivo. Here, we determined which form of K8 contains significant tumor cytotoxicity and examined how this form regulates AR expression in PCa cells and xenografts. METHODS We chose the androgen-dependent human PCa cell line LNCaP and the androgen-independent cell lines DU145 and PC-3 to study the anti-tumor potency and AR regulation mediated by Z-K8. We measured cell viability and used flow cytometry, RT-PCR, and Western blotting. Growth inhibition in vivo was evaluated with an LNCaP xenograft animal model. RESULTS In LNCaP cells, Z-K8 significantly repressed cell proliferation, induced apoptosis, repressed AR mRNA and protein expression in a time-dependent manner, and induced JNK phosphorylation. Furthermore, treatment with a JNK inhibitor significantly abolished Z-K8-induced AR downregulation. Z-K8 did not significantly inhibit reporter gene expression of constructs containing the AR promoter when it contained a mutated Sp1 binding site. Z-K8 also showed anti-tumor effects in the xenograft animal model. CONCLUSION Z-K8 not only induced LNCaP apoptosis but also reduced AR expression. These results indicate that Z-K8 may be a potential anti-tumor drug for PCa therapy.
Collapse
Affiliation(s)
- Po-Yen Liu
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Isaacs JT, D'Antonio JM, Chen S, Antony L, Dalrymple SP, Ndikuyeze GH, Luo J, Denmeade SR. Adaptive auto-regulation of androgen receptor provides a paradigm shifting rationale for bipolar androgen therapy (BAT) for castrate resistant human prostate cancer. Prostate 2012; 72:1491-505. [PMID: 22396319 PMCID: PMC3374010 DOI: 10.1002/pros.22504] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 02/06/2012] [Indexed: 01/29/2023]
Abstract
Cell culture/xenograft and gene arrays of clinical material document that development of castration resistant prostate cancer (CRPC) cells involves acquisition of adaptive auto-regulation resulting in >25-fold increase in Androgen Receptor (AR) protein expression in a low androgen environment. Such adaptive AR increase paradoxically is a liability in castrated hosts, however, when supraphysiologic androgen is acutely replaced. Cell synchronization/anti-androgen response is due to AR binding to replication complexes (RC) at origin of replication sites (ORS) in early G1 associated with licensing/restricting DNA for single round of duplication during S-phase. When CRPC cells are acutely exposed to supraphysiologic androgen, adaptively increased nuclear AR is over-stabilized, preventing sufficient degradation in mitosis, inhibiting DNA re-licensing, and thus death in the subsequent cell cycle. These mechanistic results and the fact that AR/RC binding occurs in metastatic CRPCs directly from patients provides a paradigm shifting rationale for bipolar androgen therapy (BAT) in patient progressing on chronic androgen ablation. BAT involves giving sequential cycles alternating between periods of acute supraphysiologic androgen followed by acute ablation to take advantage of vulnerability produced by adaptive auto-regulation and binding of AR to RC in CRPC cells. BAT therapy is effective in xenografts and based upon positive results has entered clinical testing.
Collapse
Affiliation(s)
- John T Isaacs
- The Chemical Therapeutic Program, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|