1
|
Bhoir S, De Benedetti A. Beyond the Horizon: Rethinking Prostate Cancer Treatment Through Innovation and Alternative Strategies. Cancers (Basel) 2024; 17:75. [PMID: 39796704 PMCID: PMC11718964 DOI: 10.3390/cancers17010075] [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: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
For nearly a century, fundamental observations that prostate cancer (PCa) cells nearly always require AR stimulation for sustained proliferation have led to a unidirectional quest to abrogate such a pathway. Similarly focused have been efforts to understand AR-driven processes in the context of elevated expression of its target genes, and much less so on products that become overexpressed when AR signaling is suppressed. Treatment with ARSI results in an increased expression of the TLK1B splice variant via a 'translational' derepression driven by the compensatory mTOR activation and consequent activation of the TLK1 > NEK1 > ATR > Chk1 and NEK1 > YAP axes. In due course, this results first in a pro-survival quiescence and then adaptation to ADT and CRPC progression. This constitutes a novel liability for PCa that we have targeted for several years and novel approaches.
Collapse
Affiliation(s)
- Siddhant Bhoir
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA 71103, USA;
- Department of Therapeutic Radiology, School of Medicine, Yale University, 15 York Street, New Haven, CT 06510, USA
| | - Arrigo De Benedetti
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA 71103, USA;
| |
Collapse
|
2
|
Kapic A, Zaman K, Nguyen V, Prokai-Tatrai K, Prokai L. Identification of Estrogen-Responsive Proteins in Mouse Seminal Vesicles Through Mass Spectrometry-Based Proteomics. Pharmaceuticals (Basel) 2024; 17:1508. [PMID: 39598420 PMCID: PMC11597337 DOI: 10.3390/ph17111508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Although estrogenic compounds promise therapeutic potential in treating various conditions, concerns regarding their endocrine-disrupting effects have been raised. Current methodologies for screening estrogenicity in rodent models are limited to the female-specific uterotrophic bioassay. Studies have reported enlargement of the seminal vesicles in orchiectomized males treated with estrogens. However, identifying estrogenicity strictly through changes in wet weights is uninformative regarding the molecular mechanisms of these agents. Therefore, protein-based biomarkers can complement and improve the sensitivity of weight-based assessments. To this end, we present a discovery-driven proteomic analysis of 17β-estradiol's effects on the seminal vesicles. Methods: We treated orchidectomized mice with the hormone for five days and used the vehicle-treated group as a control. Seminal vesicles were analyzed by shotgun approach using data-dependent nanoflow liquid chromatography-tandem mass spectrometry and label-free quantification. Proteins found to be differentially expressed between the two groups were processed through a bioinformatics pipeline focusing on pathway analyses and assembly of protein interaction networks. Results: Out of 668 identified proteins that passed rigorous validation criteria, 133 were regulated significantly by 17β-estradiol. Ingenuity Pathway Analysis® linked them to several hormone-affected pathways, including those associated with immune function such as neutrophil degranulation. The altered protein interaction networks were also related to functions including endocrine disruption, abnormal metabolism, and therapeutic effects. Conclusions: We identified several potential biomarkers for estrogenicity in mouse seminal vesicles, many of them not previously linked with exogenous 17β-estradiol exposure.
Collapse
Affiliation(s)
| | | | | | | | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.K.); (K.Z.); (V.N.); (K.P.-T.)
| |
Collapse
|
3
|
El Omari N, Khalid A, Makeen HA, Alhazmi HA, Albratty M, Mohan S, Tan CS, Ming LC, Chook JB, Bouyahya A. Stochasticity of anticancer mechanisms underlying clinical effectiveness of vorinostat. Heliyon 2024; 10:e33052. [PMID: 39021957 PMCID: PMC11253278 DOI: 10.1016/j.heliyon.2024.e33052] [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: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
The Food and Drug Administration (FDA) has approved vorinostat, also called Zolinza®, for its effectiveness in fighting cancer. This drug is a suberoyl-anilide hydroxamic acid belonging to the class of histone deacetylase inhibitors (HDACis). Its HDAC inhibitory potential allows it to accumulate acetylated histones. This, in turn, can restore normal gene expression in cancer cells and activate multiple signaling pathways. Experiments have proven that vorinostat induces histone acetylation and cytotoxicity in many cancer cell lines, increases the level of p21 cell cycle proteins, and enhances pro-apoptotic factors while decreasing anti-apoptotic factors. Additionally, it regulates the immune response by up-regulating programmed death-ligand 1 (PD-L1) and interferon gamma receptor 1 (IFN-γR1) expression, and can impact proteasome and/or aggresome degradation, endoplasmic reticulum function, cell cycle arrest, apoptosis, tumor microenvironment remodeling, and angiogenesis inhibition. In this study, we sought to elucidate the precise molecular mechanism by which Vorinostat inhibits HDACs. A deeper understanding of these mechanisms could improve our understanding of cancer cell abnormalities and provide new therapeutic possibilities for cancer treatment.
Collapse
Affiliation(s)
- Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum, 11111, Sudan
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ching Siang Tan
- School of Pharmacy, KPJ Healthcare University, Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Jack Bee Chook
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco
| |
Collapse
|
4
|
Yu Z, Peng Y, Gao J, Zhou M, Shi L, Zhao F, Wang C, Tian X, Feng L, Huo X, Zhang B, Liu M, Fang D, Ma X. The p23 co-chaperone is a succinate-activated COX-2 transcription factor in lung adenocarcinoma tumorigenesis. SCIENCE ADVANCES 2023; 9:eade0387. [PMID: 37390202 PMCID: PMC10313168 DOI: 10.1126/sciadv.ade0387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 05/30/2023] [Indexed: 07/02/2023]
Abstract
P23, historically known as a heat shock protein 90 (HSP90) co-chaperone, exerts some of its critical functions in an HSP90-independent manner, particularly when it translocates into the nucleus. The molecular nature underlying how this HSP90-independent p23 function is achieved remains as a biological mystery. Here, we found that p23 is a previously unidentified transcription factor of COX-2, and its nuclear localization predicts the poor clinical outcomes. Intratumor succinate promotes p23 succinylation at K7, K33, and K79, which drives its nuclear translocation for COX-2 transcription and consequently fascinates tumor growth. We then identified M16 as a potent p23 succinylation inhibitor from 1.6 million compounds through a combined virtual and biological screening. M16 inhibited p23 succinylation and nuclear translocation, attenuated COX-2 transcription in a p23-dependent manner, and markedly suppressed tumor growth. Therefore, our study defines p23 as a succinate-activated transcription factor in tumor progression and provides a rationale for inhibiting p23 succinylation as an anticancer chemotherapy.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Yulin Peng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Meirong Zhou
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Shi
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Feng Zhao
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Chao Wang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiangge Tian
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Feng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiaokui Huo
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Baojing Zhang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Min Liu
- Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian 116021, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaochi Ma
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
5
|
Jiang W, Wei Q, Xie H, Wu D, He H, Lv X. Effect of PTGES3 on the Prognosis and Immune Regulation in Lung Adenocarcinoma. Anal Cell Pathol (Amst) 2023; 2023:4522045. [PMID: 37416927 PMCID: PMC10322580 DOI: 10.1155/2023/4522045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/10/2023] [Accepted: 05/04/2023] [Indexed: 07/08/2023] Open
Abstract
Background PTGES3 is upregulated in multiple cancer types and promotes tumorigenesis and progression. However, the clinical outcome and immune regulation of PTGES3 in lung adenocarcinoma (LUAD) are not fully understood. This study aimed to explore the expression level and prognostic value of PTGES3 and its correlation with potential immunotherapy in LUAD. Methods All data were obtained from several databases, including the Cancer Genome Atlas database. Firstly, gene and protein expression of PTGES3 were analyzed using Tumor Immune Estimation Resource (TIMER), R software, Clinical Proteomic Tumor Analysis Consortium (CPTAC), and Human Protein Atlas (HPA). Thereafter, survival analysis was conducted using the R software, Gene Expression Profiling Interactive Analysis 2 (GEPIA2), and Kaplan-Meier Plotter. In addition, gene alteration and mutation analyses were conducted using the cBio Cancer Genomics Portal (cBioPortal) and Catalog of Somatic Mutations in Cancer (COSMIC) databases. The molecular mechanisms associated with PTGES3 were assessed via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), GeneMANIA, GEPIA2, and R software. Lastly, the role of PTGES3 in immune regulation in LUAD was investigated using TIMER, Tumor-Immune System Interaction Database (TISIDB), and SangerBox. Results The gene and protein expression of PTGES3 were elevated in LUAD tissues and compared to the normal tissues, and the high expression of PTGES3 was correlated with cancer stage and tumor grade. Survival analysis revealed that overexpression of PTGES3 was associated with poor prognosis of LUAD patients. Moreover, gene alteration and mutation analysis revealed the occurrence of several types of PTGES3 gene alterations in LUAD. Moreover, co-expression analysis and cross-analysis revealed that three genes, including CACYBP, HNRNPC, and TCP1, were correlated and interacted with PTGES3. Functional analysis of these genes revealed that PTGES3 was primarily enriched in oocyte meiosis, progesterone-mediated oocyte maturation, and arachidonic acid metabolism pathways. Furthermore, we found that PTGES3 participated in a complex immune regulation network in LUAD. Conclusion The current study indicated the crucial role of PTGES3 in LUAD prognosis and immune regulation. Altogether, our results suggested that PTGES3 could serve as a promising therapeutic and prognosis biomarker for the LUAD.
Collapse
Affiliation(s)
- Wenyan Jiang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Qiong Wei
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Haiqin Xie
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Dandan Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Haiyan He
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Xuedong Lv
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
6
|
Backe SJ, Woodford MR, Ahanin E, Sager RA, Bourboulia D, Mollapour M. Impact of Co-chaperones and Posttranslational Modifications Toward Hsp90 Drug Sensitivity. Subcell Biochem 2023; 101:319-350. [PMID: 36520312 PMCID: PMC10077965 DOI: 10.1007/978-3-031-14740-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Posttranslational modifications (PTMs) regulate myriad cellular processes by modulating protein function and protein-protein interaction. Heat shock protein 90 (Hsp90) is an ATP-dependent molecular chaperone whose activity is responsible for the stabilization and maturation of more than 300 client proteins. Hsp90 is a substrate for numerous PTMs, which have diverse effects on Hsp90 function. Interestingly, many Hsp90 clients are enzymes that catalyze PTM, demonstrating one of the several modes of regulation of Hsp90 activity. Approximately 25 co-chaperone regulatory proteins of Hsp90 impact structural rearrangements, ATP hydrolysis, and client interaction, representing a second layer of influence on Hsp90 activity. A growing body of literature has also established that PTM of these co-chaperones fine-tune their activity toward Hsp90; however, many of the identified PTMs remain uncharacterized. Given the critical role of Hsp90 in supporting signaling in cancer, clinical evaluation of Hsp90 inhibitors is an area of great interest. Interestingly, differential PTM and co-chaperone interaction have been shown to impact Hsp90 binding to its inhibitors. Therefore, understanding these layers of Hsp90 regulation will provide a more complete understanding of the chaperone code, facilitating the development of new biomarkers and combination therapies.
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Elham Ahanin
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
7
|
Labrecque MP, Alumkal JJ, Coleman IM, Nelson PS, Morrissey C. The heterogeneity of prostate cancers lacking AR activity will require diverse treatment approaches. Endocr Relat Cancer 2021; 28:T51-T66. [PMID: 33792558 PMCID: PMC8292199 DOI: 10.1530/erc-21-0002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
The use of androgen deprivation therapy and second-line anti-androgens in prostate cancer has led to the emergence of tumors employing multiple androgen receptor (AR)-dependent and AR-independent mechanisms to resist AR-targeted therapies in castration-resistant prostate cancer (CRPC). While the AR signaling axis remains the cornerstone for therapeutic development in CRPC, a clearer understanding of the heterogeneous biology of CRPC tumors is needed for innovative treatment strategies. In this review, we discuss the characteristics of CRPC tumors that lack AR activity and the temporal and spatial considerations for the conversion of an AR-dependent to an AR-independent tumor type. We describe the more prevalent treatment-emergent phenotypes arising in the CRPC disease continuum, including amphicrine, AR-low, double-negative, neuroendocrine and small cell phenotypes. We discuss the association between the loss of AR activity and tumor plasticity with a focus on the roles of transcription factors like SOX2, DNA methylation, alternative splicing, and the activity of epigenetic modifiers like EZH2, BRD4, LSD1, and the nBAF complex in conversion to a neuroendocrine or small cell phenotype in CRPC. We hypothesize that only a subset of CRPC tumors have the propensity for tumor plasticity and conversion to the neuroendocrine phenotype and outline how we might target these plastic and emergent phenotypes in CRPC. In conclusion, we assess the current and future avenues for treatment and determine that the heterogeneity of CRPCs lacking AR activity will require diverse treatment approaches.
Collapse
Affiliation(s)
- Mark P. Labrecque
- Department of Urology, University of Washington School of
Medicine, Seattle, Washington, United States of America
| | - Joshi J. Alumkal
- Department of Internal Medicine, Rogel Cancer Center,
University of Michigan, Ann Arbor, MI USA
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research
Center, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research
Center, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred Hutchinson Cancer
Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of
Medicine, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington School of
Medicine, Seattle, Washington, United States of America
- Corresponding author Telephone: 206-543-1461, Fax:
206-543-1146,
| |
Collapse
|
8
|
Eighty Years of Targeting Androgen Receptor Activity in Prostate Cancer: The Fight Goes on. Cancers (Basel) 2021; 13:cancers13030509. [PMID: 33572755 PMCID: PMC7865914 DOI: 10.3390/cancers13030509] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Prostate cancer is the second most common cancer in men world-wide, with nearly 1.3 million new cases each year, and over the next twenty years the incidence and death rate are predicted to nearly double. For decades, this lethal disease has been more or less successfully treated using hormonal therapy, which has the ultimate aim of inhibiting androgen signalling. However, prostate tumours can evade such hormonal therapies in a number of different ways and therapy resistant disease, so-called castration-resistant prostate cancer (CRPC) is the major clinical problem. Somewhat counterintuitively, the androgen receptor remains a key therapy target in CRPC. Here, we explain why this is the case and summarise both new hormone therapy strategies and the recent advances in knowledge of androgen receptor structure and function that underpin them. Abstract Prostate cancer (PCa) is the most common cancer in men in the West, other than skin cancer, accounting for over a quarter of cancer diagnoses in US men. In a seminal paper from 1941, Huggins and Hodges demonstrated that prostate tumours and metastatic disease were sensitive to the presence or absence of androgenic hormones. The first hormonal therapy for PCa was thus castration. In the subsequent eighty years, targeting the androgen signalling axis, where possible using drugs rather than surgery, has been a mainstay in the treatment of advanced and metastatic disease. Androgens signal via the androgen receptor, a ligand-activated transcription factor, which is the direct target of many such drugs. In this review we discuss the role of the androgen receptor in PCa and how the combination of structural information and functional screenings is continuing to be used for the discovery of new drug to switch off the receptor or modify its function in cancer cells.
Collapse
|
9
|
Bakir WAI, Gaidan HA, Al-Kaabi MM. Immunohistochemical expression of interlukin10 (IL10) and heat shock protein-90 (HSP-90) in prostatic carcinoma. INDIAN J PATHOL MICR 2021; 63:230-234. [PMID: 32317521 DOI: 10.4103/ijpm.ijpm_460_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background Specific cytokines are related to pathologically changed prostate, propose that the balance in cytokine differs in normal and pathological prostate. Of these cytokines the interleukins 10, due to its "pleiotropic" actions in inflammation and angiogenesis, and HSP-90 due to its expression in tumor cells at high levels, suggesting that it has an important role for growth and/or survival of tumor cells. Aims Evaluation of HSP-90 and IL10 immunoreactivity in benign prostatic hyperplasia (BPH) and prostatic carcinoma and to correlate this expression with clinicopathological parameters. Settings and Design A retrospective study in which 83 Paraffin-embedded tissue specimens including (43) BPH, (40) prostatic carcinoma and (20) normal prostate as control were included between the period of January 2015 and January 2017. Patients, Material and Methods All the cases were evaluated histopathologically and stained immunohistochemically for IL10 and HSP-90. Only cytoplasmic staining was considered as positive. Immunoreactivity scoring for both markers expression was calculated based on both staining intensity and percentage. Statistical Analysis Was done using SPSS Version 21 statistical analysis software. P value of <0.05 was considered statistically significant. Result Statistical analysis of HSP-90 and IL10 expression revealed a highly significant correlation of expression of these two markers in advanced Gleason grading and tumor, node, and metastasis (TNM) staging cases of prostatic carcinoma. Conclusion High expression of IL10 and HSP-90 is associated with high grade and stage of prostatic carcinoma. This provides a base for further studies and researches on the role of these investigated proteins as prognostic markers immunotherapy targets for carcinoma of the prostate.
Collapse
Affiliation(s)
- Wasan Abdul-Ilah Bakir
- Department of Microbiology, Mustansiriyah University, College of Medicine, Baghdad, Iraq
| | - Hiba Ahmed Gaidan
- Department of Pathology, Mustansiriyah University, College of Medicine, Baghdad, Iraq
| | - Methaq Mueen Al-Kaabi
- Department of Pathology, Mustansiriyah University, College of Medicine, Baghdad, Iraq
| |
Collapse
|
10
|
Ye M, Tian H, Lin S, Mo J, Li Z, Chen X, Liu J. Resveratrol inhibits proliferation and promotes apoptosis via the androgen receptor splicing variant 7 and PI3K/AKT signaling pathway in LNCaP prostate cancer cells. Oncol Lett 2020; 20:169. [PMID: 32934736 PMCID: PMC7471767 DOI: 10.3892/ol.2020.12032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is a common malignant tumor of the male genitourinary system and its incidence increases with age. Studies have shown that resveratrol (Res) inhibits cancer cell proliferation, migration, invasion and promotes apoptosis. The present study evaluated the effect of Res in two human prostate cancer cell lines (the androgen-dependent LNCaP cell line and the non-androgen-independent LNCaP-B cell line) on proliferation and apoptosis. A proliferation assay was used to demonstrate that Res inhibited proliferation of LNCaP and LNCaP-B cells in the range of 25-100 µM, and the effect was time- and dose-dependent. Using flow cytometry, it was reported that various concentrations of Res induced apoptosis in LNCaP and LNCaP-B cells, and that the apoptotic effect of Res was dose-dependent. A chemiluminescence assay showed that Res inhibited prostate specific antigen levels in LNCaP and LNCaP-B cells. Reverse transcription quantitative-PCR showed that Res inhibited the expression of androgen receptor (AR) in LNCaP and LNCaP-B cells at the mRNA level. Western blot analysis showed that Res suppressed the expression of AR protein as well as protein kinase B (AKT) phosphorylation. To study the effect of Res on the expression of AR splicing variant 7 (ARV7) and the PI3K/AKT signaling pathway in prostate cancer cells, as well as the underlying molecular mechanisms, the recombinant ARV7 expression vector Pcdna3.1-ARV7 was transfected into LNCaP and LNCaP cells and the aforementioned experiments were repeated. It was revealed that Res acted via the ARV7 and the AKT pathways. Taken together, the present results suggested that Res suppresses the proliferation of prostate cancer cells, promotes apoptosis and inhibits the expression of AR mRNA and protein. These effects likely resulted from inhibition of ARV7 and the AKT signaling pathway.
Collapse
Affiliation(s)
- Mushi Ye
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Huanshu Tian
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Shanhong Lin
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jierong Mo
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Zhuo Li
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Xiaojun Chen
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| |
Collapse
|
11
|
Hoter A, Rizk S, Naim HY. The Multiple Roles and Therapeutic Potential of Molecular Chaperones in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11081194. [PMID: 31426412 PMCID: PMC6721600 DOI: 10.3390/cancers11081194] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common cancer types in men worldwide. Heat shock proteins (HSPs) are molecular chaperones that are widely implicated in the pathogenesis, diagnosis, prognosis, and treatment of many cancers. The role of HSPs in PCa is complex and their expression has been linked to the progression and aggressiveness of the tumor. Prominent chaperones, including HSP90 and HSP70, are involved in the folding and trafficking of critical cancer-related proteins. Other members of HSPs, including HSP27 and HSP60, have been considered as promising biomarkers, similar to prostate-specific membrane antigen (PSMA), for PCa screening in order to evaluate and monitor the progression or recurrence of the disease. Moreover, expression level of chaperones like clusterin has been shown to correlate directly with the prostate tumor grade. Hence, targeting HSPs in PCa has been suggested as a promising strategy for cancer therapy. In the current review, we discuss the functions as well as the role of HSPs in PCa progression and further evaluate the approach of inhibiting HSPs as a cancer treatment strategy.
Collapse
Affiliation(s)
- Abdullah Hoter
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sandra Rizk
- School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Hassan Y Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| |
Collapse
|
12
|
Bharadwaj S, Lee KE, Dwivedi VD, Yadava U, Kang SG. Computational aided mechanistic understanding of Camellia sinensis bioactive compounds against co-chaperone p23 as potential anticancer agent. J Cell Biochem 2019; 120:19064-19075. [PMID: 31257629 DOI: 10.1002/jcb.29229] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 11/08/2022]
Abstract
Co-chaperon p23 has been well established as molecular chaperon for the heat shock protein 90 (Hsp90) that further leads to immorality in cancer cells by providing defense against Hsp90 inhibitors, and as stimulating agent for generating overexpressed antiapoptotic proteins, that is, Hsp70 and Hsp27. The natural compounds such as catechins from Camellia sinensis (green tea) are also well known for inhibition activity against various cancer. However, molecular interaction profile and potential lead bioactive compounds against co-chaperon p23 from green tea are not yet reported. To this context, we study the various secondary metabolites of green tea against co-chaperon p23 using structure-based virtual screening from Traditional Chinese Medicine (TCM) database. Following 26 compounds were obtained from TCM database and further studied for extra precision molecular docking that showed binding score between -10.221 and -2.276 kcal/mol with co-chaperon p23. However, relative docking score to known inhibitors, that is, ailanthone (-4.54 kcal/mol) and gedunin ( 3.60 kcal/mol) along with ADME profile analysis concluded epicatechin (-7.013 kcal/mol) and cis-theaspirone (-4.495 kcal/mol) as potential lead inhibitors from green tea against co-chaperone p23. Furthermore, molecular dynamics simulation and molecular mechanics generalized born surface area calculations validated that epicatechin and cis-theaspirone have significantly occupied the active region of co-chaperone p23 by hydrogen and hydrophobic interactions with various residues including most substantial amino acids, that is, Thr90, Ala94, and Lys95. Hence, these results supported the fact that green tea contained potential compounds with an ability to inhibit the cancer by disrupting the co-chaperon p23 activity.
Collapse
Affiliation(s)
- Shiv Bharadwaj
- Department of Biotechnology, Institute of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Kyung Eun Lee
- Department of Biotechnology, Institute of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Vivek Dhar Dwivedi
- Centre for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida, India
| | - Umesh Yadava
- Department of Physics, Deen Dayal Upadhyay Gorakhpur University, Gorakhpur, India
| | - Sang Gu Kang
- Department of Biotechnology, Institute of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea.,Stemforce, 313 Institute of Industrial Technology, Yeungnam University, Gyeongbuk, Gyeongsan, Republic of Korea
| |
Collapse
|
13
|
Rodríguez-Blanco G, Zeneyedpour L, Duijvesz D, Hoogland AM, Verhoef EI, Kweldam CF, Burgers PC, Smitt PS, Bangma CH, Jenster G, van Leenders GJLH, Dekker LJM, Luider TM. Tissue proteomics outlines AGR2 AND LOX5 as markers for biochemical recurrence of prostate cancer. Oncotarget 2018; 9:36444-36456. [PMID: 30559929 PMCID: PMC6284859 DOI: 10.18632/oncotarget.26342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/21/2018] [Indexed: 12/22/2022] Open
Abstract
Although many patients are cured from prostate cancer (PCa) by surgery only, there are still patients who will experience rising prostate-specific antigen (PSA) levels after surgery, a condition known as biochemical recurrence (BCR). Novel protein prognostic markers in PCa tissue might enable finding better treatment for those patients experiencing BCR with a high chance of metastasis. In this study, we aimed to identify altered proteins in prostate cancer tissue, and to evaluate their potential role as prognostic markers. We used two proteomics strategies to analyse 34 prostate tumours (PCa) and 33 normal adjacent prostate (NAP) tissues. An independent cohort of 481 samples was used to evaluate the expression of three proteins: AGR2, FASN and LOX5 as prognostic markers of the disease. Tissue microarray immunohistochemical staining indicated that a low percentage of positive tumour cells for AGR2 (HR (95% CI) = 0.61 (0.43-0.93)), and a low percentage of positive tumour cells for LOX5 expression (HR (95% CI) = 2.53 (1.23-5.22)) are predictors of BCR after RP. In contrast, FASN expression had no prognostic value for PCa.
Collapse
Affiliation(s)
| | - Lona Zeneyedpour
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Diederick Duijvesz
- Department of Urology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - A Marije Hoogland
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Peter C Burgers
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Chris H Bangma
- Department of Urology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Guido Jenster
- Department of Urology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Lennard J M Dekker
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Theo M Luider
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
14
|
p23 protects the human aryl hydrocarbon receptor from degradation via a heat shock protein 90-independent mechanism. Biochem Pharmacol 2018; 152:34-44. [PMID: 29555469 DOI: 10.1016/j.bcp.2018.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/14/2018] [Indexed: 01/29/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated signaling molecule which is involved in diverse biological functions ranging from cancer metastasis to immune regulation. This receptor forms a cytoplasmic complex with Hsp90, p23, and XAP2. We have previously reported that down-regulation of p23 triggers degradation of the AHR protein, uncovering a potentially dynamic event which controls the cellular AHR levels without ligand treatment. Here we investigate the underlying mechanisms for this p23 effect using wild-type HeLa and the p23 knockdown HeLa cells. Reduction of the Hsp90 and XAP2 contents, however, did not affect the AHR protein levels, implying that this p23 effect on AHR is more than just alteration of the cytoplasmic complex dynamics. Association of p23 with Hsp90 is not important for the modulation of the AHR levels since exogenous expression of p23 mutants with modest Hsp90-binding affinity effectively restored the AHR message and protein levels. The protein folding property of p23 which resides at the terminal 50-amino acid region is not involved for this p23 effect. Results from our interaction study using the affinity purified thioredoxin fusion proteins and GST fusion proteins showed that p23 directly interacts with AHR and the interaction surface lies within AHR amino acid 1-216 and p23 amino acid 1-110. Down-regulation of the p23 protein content promotes the ubiquitination of AHR, indicating that p23 protects AHR from the ubiquitin-meditated protein degradation.
Collapse
|
15
|
Hsp90 Co-chaperone p23 contributes to dopaminergic mitochondrial stress via stabilization of PHD2: Implications for Parkinson's disease. Neurotoxicology 2018; 65:166-173. [PMID: 29471019 DOI: 10.1016/j.neuro.2018.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 12/17/2022]
Abstract
The heat shock factor 90 (hsp90) complex has long been associated with neuropathological phenotypes linked to Parkinson's disease (PD) and its inhibition is neuroprotective in disease models. Hsp90 is conventionally believed to act by suppressing induction of hsp70. Here, we report a novel hsp70-independent mechanism by which Hsp90 may also contribute to PD-associated neuropathology. We previously reported that inhibition of the enzyme prolyl hydroxylase domain 2 (PHD2) in conjunction with increases in hypoxia-inducible factor 1 alpha (HIF1α) results in protection of vulnerable dopaminergic substantia nigra pars compacta (DAergic SNpc) neurons in in vitro and in vivo models of PD. We discovered an increased interaction between PHD2 and the p23:Hsp90 chaperone complex in response to mitochondrial stress elicited by the mitochondrial neurotoxin 1-methyl-4-phenylpyridine (MPP+) within cultured DAergic cells. Genetic p23 knockdown was found to result in decreases in steady-state PHD2 protein and activity and reduced susceptibility to MPP+ neurotoxicity. Administration of the p23 inhibitor gedunin was also neuroprotective in these cells as well as in human induced pluripotent stem cell (iPSC)-derived neurons. Our data suggests that mitochondrial stress-mediated elevations in PHD2 interaction with the p23-hsp90 complex have detrimental effects on the survival of DAergic neurons, while p23 inhibition is neuroprotective. We propose that neurotoxic effects are tied to enhanced PHD2 stabilization by the hsp90-p23 chaperone complex that is abrogated by p23 inhibition. This demonstrates a novel connection between two independent pathways previously linked to PD, hsp90 and PHD2-HIF1α, which could have important implications for here-to-fore unexplored mechanisms underlying PD neuropathology.
Collapse
|
16
|
The histone deacetylase inhibitor SAHA induces HSP60 nitration and its extracellular release by exosomal vesicles in human lung-derived carcinoma cells. Oncotarget 2018; 7:28849-67. [PMID: 26700624 PMCID: PMC5045361 DOI: 10.18632/oncotarget.6680] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/22/2015] [Indexed: 12/14/2022] Open
Abstract
HSP60 undergoes changes in quantity and distribution in some types of tumors suggesting a participation of the chaperonin in the mechanism of transformation and cancer progression. Suberoylanilide hydroxamic acid (SAHA), a member of a family of histone deacetylase inhibitors (HDACi), has anti-cancer potential but its interaction, if any, with HSP60 has not been elucidated. We investigated the effects of SAHA in a human lung-derived carcinoma cell line (H292). We analysed cell viability and cycle; oxidative stress markers; mitochondrial integrity; HSP60 protein and mRNA levels; and HSP60 post-translational modifications, and its secretion. We found that SAHA is cytotoxic for H292 cells, interrupting the cycle at the G2/M phase, which is followed by death; cytotoxicity is associated with oxidative stress, mitochondrial damage, and diminution of intracellular levels of HSP60; HSP60 undergoes a post-translational modification and becomes nitrated; and nitrated HSP60 is exported via exosomes. We propose that SAHA causes ROS overproduction and mitochondrial dysfunction, which leads to HSP60 nitration and release into the intercellular space and circulation to interact with the immune system. These successive steps might constitute the mechanism of the anti-tumor action of SAHA and provide a basis to design supplementary therapeutic strategies targeting HSP60, which would be more efficacious than the compound alone.
Collapse
|
17
|
Cha S, Shin DH, Seok JR, Myung JK. Differential proteome expression analysis of androgen-dependent and -independent pathways in LNCaP prostate cancer cells. Exp Cell Res 2017; 359:215-225. [DOI: 10.1016/j.yexcr.2017.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022]
|
18
|
Defective glucocorticoid receptor signaling and keratinocyte-autonomous defects contribute to skin phenotype of mouse embryos lacking the Hsp90 co-chaperone p23. PLoS One 2017. [PMID: 28650975 PMCID: PMC5484504 DOI: 10.1371/journal.pone.0180035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
p23 is a small acidic protein with intrinsic molecular chaperone activity. It is best known as a co-chaperone of the major cytosolic molecular chaperone Hsp90. p23 binds the N-terminus of Hsp90 and stabilizes the ATP-bound and N-terminally closed Hsp90 dimer. It is in this configuration that many Hsp90 clients are most stably bound. Considering the important role of p23 in the Hsp90 cycle, it came as a surprise that it is not absolutely essential for viability in the budding yeast or for mouse development. Mice without p23 develop quite normally until birth and then all die perinatally because of immature lungs. The only other apparent phenotype of late stage embryos and newborns is a skin defect, which we have further characterized here. We found that skin differentiation is impaired, and that both apoptosis and cell proliferation are augmented in the absence of p23; the consequences are a severe thinning of the stratum corneum and reduced numbers of hair follicles. The altered differentiation, spontaneous apoptosis and proliferation are all mimicked by isolated primary keratinocytes indicating that they do require p23 functions in a cell-autonomous fashion. Since the phenotype of p23-null embryos is strikingly similar to that of embryos lacking the glucocorticoid receptor, a paradigmatic Hsp90-p23 client protein, we investigated glucocorticoid signaling. We discovered that it is impaired in vivo and for some aspects in isolated keratinocytes. Our results suggest that part of the phenotype of p23-null embryos can be explained by an impact on this particular Hsp90 client, but do not exclude that p23 by itself or in association with Hsp90 affects skin development and homeostasis through yet other pathways.
Collapse
|
19
|
Ailanthone targets p23 to overcome MDV3100 resistance in castration-resistant prostate cancer. Nat Commun 2016; 7:13122. [PMID: 27959342 PMCID: PMC5159881 DOI: 10.1038/ncomms13122] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/02/2016] [Indexed: 01/17/2023] Open
Abstract
Androgen receptor (AR) antagonist MDV3100 is the first therapeutic approach in treating castration-resistant prostate cancer (CRPC), but tumours frequently become drug resistant via multiple mechanisms including AR amplification and mutation. Here we identify the small molecule Ailanthone (AIL) as a potent inhibitor of both full-length AR (AR-FL) and constitutively active truncated AR splice variants (AR-Vs). AIL binds to the co-chaperone protein p23 and prevents AR's interaction with HSP90, thus resulting in the disruption of the AR-chaperone complex followed by ubiquitin/proteasome-mediated degradation of AR as well as other p23 clients including AKT and Cdk4, and downregulates AR and its target genes in PCa cell lines and orthotopic animal tumours. In addition, AIL blocks tumour growth and metastasis of CRPC. Finally, AIL possesses favourable drug-like properties such as good bioavailability, high solubility, lack of CYP inhibition and low hepatotoxicity. In general, AIL is a potential candidate for the treatment of CRPC. Prostate cancers often become castration resistant due to alternative expression of androgen receptor (AR) splice variants. Here, the authors screened a library of natural compounds and identified Ailanthone as a potent inhibitor of AR through its binding to the co-chaperone protein p23 that, by preventing AR interaction with HSP90, results in ubiquitin/proteasome-mediated degradation of the receptor.
Collapse
|
20
|
Abstract
Prostate cancer (PCa) is one of the most lethal cancers in western countries. Androgen receptor (AR) signaling pathway plays a key role in PCa progression. Despite the initial effectiveness of androgen deprivation therapy (ADT)for treatment of patients with advanced PCa, most of them will develop resistance to ADT and progress to metastatic castration resistant prostate cancer (mCRPC). Constitutively transcriptional activated AR splice variants (AR-Vs) have emerged as critical players in the development and progression of mCRPC. Among AR-Vs identified to date, AR-V7 (a.k.a. AR3) is one of the most abundant and frequently found in both PCa cell lines and in human prostate tissues. Most of functional studies have been focused on AR-V7/AR3 and revealed its role in regulation of survival, growth, differentiation and migration in prostate cells. In this review, we will summarize our current understanding of regulation of expression and activity of AR-Vs in mCRPC.
Collapse
Affiliation(s)
- Jin Xu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yun Qiu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Foley C, Mitsiades N. Moving Beyond the Androgen Receptor (AR): Targeting AR-Interacting Proteins to Treat Prostate Cancer. HORMONES & CANCER 2016; 7:84-103. [PMID: 26728473 PMCID: PMC5380740 DOI: 10.1007/s12672-015-0239-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
Medical or surgical castration serves as the backbone of systemic therapy for advanced and metastatic prostate cancer, taking advantage of the importance of androgen signaling in this disease. Unfortunately, resistance to castration emerges almost universally. Despite the development and approval of new and more potent androgen synthesis inhibitors and androgen receptor (AR) antagonists, prostate cancers continue to develop resistance to these therapeutics, while often maintaining their dependence on the AR signaling axis. This highlights the need for innovative therapeutic approaches that aim to continue disrupting AR downstream signaling but are orthogonal to directly targeting the AR itself. In this review, we discuss the preclinical research that has been done, as well as clinical trials for prostate cancer, on inhibiting several important families of AR-interacting proteins, including chaperones (such as heat shock protein 90 (HSP90) and FKBP52), pioneer factors (including forkhead box protein A1 (FOXA1) and GATA-2), and AR transcriptional coregulators such as the p160 steroid receptor coactivators (SRCs) SRC-1, SRC-2, SRC-3, as well as lysine deacetylases (KDACs) and lysine acetyltransferases (KATs). Researching the effect of-and developing new therapeutic agents that target-the AR signaling axis is critical to advancing our understanding of prostate cancer biology, to continue to improve treatments for prostate cancer and for overcoming castration resistance.
Collapse
Affiliation(s)
- Christopher Foley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA
| | - Nicholas Mitsiades
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Suite R407, MS: BCM187, Houston, TX, 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Hsp90 Co-chaperones as Drug Targets in Cancer: Current Perspectives. TOPICS IN MEDICINAL CHEMISTRY 2016. [DOI: 10.1007/7355_2015_99] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
A phase II trial of ganetespib, a heat shock protein 90 Hsp90) inhibitor, in patients with docetaxel-pretreated metastatic castrate-resistant prostate cancer (CRPC)-a prostate cancer clinical trials consortium (PCCTC) study. Invest New Drugs 2015; 34:112-8. [PMID: 26581400 DOI: 10.1007/s10637-015-0307-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/09/2015] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Heat shock protein 90 (Hsp90) has been studied as a therapeutic target in many cancers. In preclinical trials, the Hsp90 ATPase inhibitor ganetespib demonstrated potent inhibition of solid tumor growth, with superior potency than prior Hsp90 inhibitors. Given the promising preclinical outcome and favorable pharmacologic properties of ganetespib, we conducted a phase II trial of single-agent ganetespib in patients with metastatic, castrate-resistant prostate cancer (mCRPC). The primary objective of the study was to determine the 6-month progression-free survival (PFS) rate. METHODS Patients with mCRPC who had been previously treated with docetaxel were enrolled after meeting eligibility criteria. All patients received ganetespib at 200 mg/m(2) on days 1, 8, and 15 of every 28 days (one cycle). Subjects who tolerated therapy were continued on ganetespib until disease progression. Considering that Hsp90 acetylation may confer insensitivity to Hsp90 inhibitors and maspin inhibits protein deacetylation, maspin-associated molecular markers were evaluated. RESULTS Eighteen patients were recruited into the trial; most were Caucasian, had performance status 1, had received prior docetaxel, and were heavily pretreated. Of the 17 patients who were treated, none attained 6-month PFS. Only 2 patients achieved PFS > 4 months. The median PFS was 1.9 months. As per the study design, the trial was terminated after the interim analysis. The most frequent types of Grade 3 toxicity were dehydration, diarrhea, and fatigue. Molecular markers provided little additional insight regarding drug activity. CONCLUSIONS Ganetespib demonstrated minimal clinical activity in men with mCRPC. The true 6-month PFS rate was, at most, 0.20. Possible reasons for this include selection of a heavily pretreated patient population and lack of agent potency in patients with mCRPC.
Collapse
|
24
|
La Colla A, Pronsato L, Milanesi L, Vasconsuelo A. 17β-Estradiol and testosterone in sarcopenia: Role of satellite cells. Ageing Res Rev 2015; 24:166-77. [PMID: 26247846 DOI: 10.1016/j.arr.2015.07.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/24/2015] [Accepted: 07/30/2015] [Indexed: 12/25/2022]
Abstract
The loss of muscle mass and strength with aging, referred to as sarcopenia, is a prevalent condition among the elderly. Although the molecular mechanisms underlying sarcopenia are unclear, evidence suggests that an age-related acceleration of myocyte loss via apoptosis might be responsible for muscle perfomance decline. Interestingly, sarcopenia has been associated to a deficit of sex hormones which decrease upon aging. The skeletal muscle ability to repair and regenerate itself would not be possible without satellite cells, a subpopulation of cells that remain quiescent throughout life. They are activated in response to stress, enabling them to guide skeletal muscle regeneration. Thus, these cells could be a key factor to overcome sarcopenia. Of importance, satellite cells are 17β-estradiol (E2) and testosterone (T) targets. In this review, we summarize potential mechanisms through which these hormones regulate satellite cells activation during skeletal muscle regeneration in the elderly. The advance in its understanding will help to the development of potential therapeutic agents to alleviate and treat sarcopenia and other related myophaties.
Collapse
|
25
|
Zhang J, Tan K, Meng X, Yang W, Wei H, Sun R, Yin L, Pu Y. Overexpression of G6PD and HSP90 Beta in Mice with Benzene Exposure Revealed by Serum Peptidome Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:11241-53. [PMID: 26378550 PMCID: PMC4586672 DOI: 10.3390/ijerph120911241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/15/2015] [Accepted: 09/01/2015] [Indexed: 12/04/2022]
Abstract
The small peptides representation of the original proteins are a valuable source of information that can be used as biomarkers involved in toxicity mechanism for chemical exposure. The aim of this study is to investigate serum peptide biomarkers of benzene exposure. C57BL/6 mice were enrolled into control group and benzene groups of 150 and 300 mg/kg/d Serum peptides were identified by mass spectrometry using an assisted laser desorption ionization/time of flight mass spectrometry (MS). Differential peptide spectra were obtained by tandem mass spectrometry and analyzed by searching the International Protein Index using the Sequest program. Forty-one peptide peaks were found in the range of 1000-10,000 Da molecular weight. Among them, seven peaks showed significantly different expression between exposure groups and control group. Two peptide peaks (1231.2 and 1241.8), which showed a two-fold increase in expression, were sequenced and confirmed as glucose 6-phosphate dehydrogenase (G6PD) and heat shock protein 90 Beta (HSP90 Beta), respectively. Furthermore, the expression of the two proteins in liver cells showed the same trend as in serum. In conclusion, G6PD and HSP90 beta might be the candidate serum biomarkers of benzene exposure. It also provided possible clues for the molecular mechanism of benzene-induced oxidative stress.
Collapse
Affiliation(s)
- Juan Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Kehong Tan
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Xing Meng
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Wenwen Yang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Haiyan Wei
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Rongli Sun
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
26
|
Cato L, Neeb A, Brown M, Cato ACB. Control of steroid receptor dynamics and function by genomic actions of the cochaperones p23 and Bag-1L. NUCLEAR RECEPTOR SIGNALING 2014; 12:e005. [PMID: 25422595 PMCID: PMC4242288 DOI: 10.1621/nrs.12005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/20/2014] [Indexed: 01/23/2023]
Abstract
Molecular chaperones encompass a group of unrelated proteins that facilitate the
correct assembly and disassembly of other macromolecular structures, which they
themselves do not remain a part of. They associate with a large and diverse set
of coregulators termed cochaperones that regulate their function and
specificity. Amongst others, chaperones and cochaperones regulate the activity
of several signaling molecules including steroid receptors, which upon ligand
binding interact with discrete nucleotide sequences within the nucleus to
control the expression of diverse physiological and developmental genes.
Molecular chaperones and cochaperones are typically known to provide the correct
conformation for ligand binding by the steroid receptors. While this
contribution is widely accepted, recent studies have reported that they further
modulate steroid receptor action outside ligand binding. They are thought to
contribute to receptor turnover, transport of the receptor to different
subcellular localizations, recycling of the receptor on chromatin and even
stabilization of the DNA-binding properties of the receptor. In addition to
these combined effects with molecular chaperones, cochaperones are reported to
have additional functions that are independent of molecular chaperones. Some of
these functions also impact on steroid receptor action. Two well-studied
examples are the cochaperones p23 and Bag-1L, which have been identified as
modulators of steroid receptor activity in nuclei. Understanding details of
their regulatory action will provide new therapeutic opportunities of
controlling steroid receptor action independent of the widespread effects of
molecular chaperones.
Collapse
Affiliation(s)
- Laura Cato
- Division of Molecular and Cellular Oncology, Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA (LC, MB) and Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany (AN, ACBC)
| | - Antje Neeb
- Division of Molecular and Cellular Oncology, Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA (LC, MB) and Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany (AN, ACBC)
| | - Myles Brown
- Division of Molecular and Cellular Oncology, Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA (LC, MB) and Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany (AN, ACBC)
| | - Andrew C B Cato
- Division of Molecular and Cellular Oncology, Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA (LC, MB) and Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany (AN, ACBC)
| |
Collapse
|
27
|
The co-chaperone p23 promotes prostate cancer motility and metastasis. Mol Oncol 2014; 9:295-308. [PMID: 25241147 PMCID: PMC4510206 DOI: 10.1016/j.molonc.2014.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer is an androgen receptor (AR)‐dependent malignancy at initiation and progression, therefore hormone therapy is the primary line of systemic treatment. Despite initial disease regression, tumours inevitably recur and progress to an advanced castration‐resistant state a major feature of which is metastasis to the bone. Up‐regulation of AR cofactors and chaperones that overcome low hormone conditions to maintain basal AR activity has been postulated as a mechanism of therapy relapse. p23, an essential component of the apo‐AR complex, acts also after ligand binding to increase AR transcriptional activity and target gene expression, partly by increasing chromatin‐loaded holo‐receptor‐complexes. Immunohistochemical studies have demonstrated increased p23 expression in advanced prostate cancer. Here, we further characterise p23 roles in AR signalling and show that it modulates cytosolic AR levels in the absence of hormone, confirming a chaperoning function in the aporeceptor complex and suggesting p23 upregulates AR signalling at multiple stages. Moreover, p23 protein levels significantly increased upon treatment with not only androgen but also clinically relevant anti‐androgens. This was in contrast to the HSP90 inhibitor 17‐AAG, which did not modulate expression of the cochaperone – important given the HSP90‐independent roles we and others have previously described for p23. Further, we demonstrate p23 is implicated in prostate cancer cell motility and in acquisition of invasiveness capacity through the expression of specific genes known to participate in cancer progression. This may drive metastatic processes in vivo since analysis of prostate tumour biopsies revealed that high nuclear p23 significantly correlated with shorter survival times and with development of metastases in patients with lower grade tumours. We propose that increased p23 expression may allow cells to acquire a more aggressive phenotype, contributing to disease progression, and that p23 is a plausible secondary target in combination with HSP90 inhibition as a potential therapy for advanced prostate cancer. We report a novel function for p23 in prostate cancer progression. p23 protein levels increase upon treatment with androgens and anti‐androgens. p23 promotes prostate cancer cell motility and acquisition of invasiveness. High nuclear p23 significantly correlates with shorter survival times in patients. p23 may reprogram gene expression profile into a more metastatic pattern.
Collapse
|
28
|
Sprenger CCT, Plymate SR. The link between androgen receptor splice variants and castration-resistant prostate cancer. Discov Oncol 2014; 5:207-17. [PMID: 24798453 DOI: 10.1007/s12672-014-0177-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 04/03/2014] [Indexed: 01/19/2023] Open
Abstract
Resistance to the latest advanced prostate cancer therapies, including abiraterone and enzalutamide, is associated with increased expression of constitutively active androgen receptor splice variants (AR-Vs). The exact mechanism by which these therapies result in AR-Vs is unknown, but may include genomic rearrangement of the androgen receptor gene as well as alternative splicing of the AR pre-messenger RNA (mRNA). An additional complication that hinders further development of effective AR strategies is that the mechanisms by which the directed therapies are bypassed may vary. Finally, the question must be addressed as to whether the androgen receptor remains to be the driver of most castration resistant disease or whether truly AR-independent tumors arise after successful androgen ablation therapy. In this review, we will examine androgen receptor splice variants as an alternative mechanism by which prostate cancer becomes resistant to androgen receptor-directed therapy.
Collapse
|
29
|
Constitutively-active androgen receptor variants function independently of the HSP90 chaperone but do not confer resistance to HSP90 inhibitors. Oncotarget 2014; 4:691-704. [PMID: 23674566 PMCID: PMC3742830 DOI: 10.18632/oncotarget.975] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The development of lethal, castration resistant prostate cancer is associated with adaptive changes to the androgen receptor (AR), including the emergence of mutant receptors and truncated, constitutively active AR variants. AR relies on the molecular chaperone HSP90 for its function in both normal and malignant prostate cells, but the requirement for HSP90 in environments with aberrant AR expression is largely unknown. Here, we investigated the efficacy of three HSP90 inhibitors, 17-AAG, HSP990 and AUY922, against clinically-relevant AR missense mutants and truncated variants. HSP90 inhibition effectively suppressed the signaling of wild-type AR and all AR missense mutants tested. By contrast, two truncated AR variants, AR-V7 and ARv567es, exhibited marked resistance to HSP90 inhibitors. Supporting this observation, nuclear localization of the truncated AR variants was not affected by HSP90 inhibition and AR variant:HSP90 complexes could not be detected in prostate cancer cells. Interestingly, HSP90 inhibition resulted in accumulation of AR-V7 and ARv567es in both cell lines and human tumor explants. Despite the apparent independence of AR variants from HSP90 and their treatment-associated induction, the growth of cell lines with endogenous or enforced expression of AR-V7 or ARv567es remained highly sensitive to AUY922. This study demonstrates that functional AR variant signaling does not confer resistance to HSP90 inhibition, yields insight into the interaction between AR and HSP90 and provides further impetus for the clinical application of HSP90 inhibitors in advanced prostate cancer.
Collapse
|
30
|
Shaheed SU, Rustogi N, Scally A, Wilson J, Thygesen H, Loizidou MA, Hadjisavvas A, Hanby A, Speirs V, Loadman P, Linforth R, Kyriacou K, Sutton CW. Identification of stage-specific breast markers using quantitative proteomics. J Proteome Res 2013; 12:5696-708. [PMID: 24106833 DOI: 10.1021/pr400662k] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Matched healthy and diseased tissues from breast cancer patients were analyzed by quantitative proteomics. By comparing proteomic profiles of fibroadenoma (benign tumors, three patients), DCIS (noninvasive cancer, three patients), and invasive ductal carcinoma (four patients), we identified protein alterations that correlated with breast cancer progression. Three 8-plex iTRAQ experiments generated an average of 826 protein identifications, of which 402 were common. After excluding those originating from blood, 59 proteins were significantly changed in tumor compared with normal tissues, with the majority associated with invasive carcinomas. Bioinformatics analysis identified relationships between proteins in this subset including roles in redox regulation, lipid transport, protein folding, and proteasomal degradation, with a substantial number increased in expression due to Myc oncogene activation. Three target proteins, cofilin-1 and p23 (increased in invasive carcinoma) and membrane copper amine oxidase 3 (decreased in invasive carcinoma), were subjected to further validation. All three were observed in phenotype-specific breast cancer cell lines, normal (nontransformed) breast cell lines, and primary breast epithelial cells by Western blotting, but only cofilin-1 and p23 were detected by multiple reaction monitoring mass spectrometry analysis. All three proteins were detected by both analytical approaches in matched tissue biopsies emulating the response observed with proteomics analysis. Tissue microarray analysis (361 patients) indicated cofilin-1 staining positively correlating with tumor grade and p23 staining with ER positive status; both therefore merit further investigation as potential biomarkers.
Collapse
Affiliation(s)
- Sadr-ul Shaheed
- Institute of Cancer Therapeutics, University of Bradford , Tumbling Hill Street, Bradford BD7 1DP, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Current World Literature. Curr Opin Urol 2013. [DOI: 10.1097/mou.0b013e3283605159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Cano LQ, Lavery DN, Bevan CL. Mini-review: Foldosome regulation of androgen receptor action in prostate cancer. Mol Cell Endocrinol 2013; 369:52-62. [PMID: 23395916 DOI: 10.1016/j.mce.2013.01.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/28/2013] [Accepted: 01/29/2013] [Indexed: 11/24/2022]
Abstract
Steroid hormone receptors play diverse roles in many aspects of human physiology including cell division, apoptosis and homeostasis, tissue differentiation, sexual development and response to stress. These ligand-activated transcription factors require the functional activity of numerous chaperone and chaperone-associated proteins, collectively termed the foldosome, at the crucial step of ligand recognition and binding. Since the initial isolation of foldosome components and pioneering research by Pratt, Toft and colleagues we understand much regarding cytosolic receptor function. The classical view, that the role of foldosome components is restricted to the cytosol, has been modified over recent years by research highlighting additional roles of chaperone proteins in nuclear translocation and target gene expression. Further, dysregulation of chaperone activity and expression has been implicated in various cancers, including breast and prostate cancer. Consequently, the foldosome provides an attractive therapeutic target in steroid hormone receptor-driven malignancies. This review summarises current knowledge of how the foldosome impacts upon androgen receptor signalling, which is the key therapeutic target on prostate cancer, and how foldosome components may be used as biomarkers or therapeutic targets in this disease.
Collapse
Affiliation(s)
- Laia Querol Cano
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
33
|
Calderwood SK. Molecular cochaperones: tumor growth and cancer treatment. SCIENTIFICA 2013; 2013:217513. [PMID: 24278769 PMCID: PMC3820307 DOI: 10.1155/2013/217513] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/01/2013] [Indexed: 05/12/2023]
Abstract
Molecular chaperones play important roles in all cellular organisms by maintaining the proteome in an optimally folded state. They appear to be at a premium in cancer cells whose evolution along the malignant pathways requires the fostering of cohorts of mutant proteins that are employed to overcome tumor suppressive regulation. To function at significant rates in cells, HSPs interact with cochaperones, proteins that assist in catalyzing individual steps in molecular chaperoning as well as in posttranslational modification and intracellular localization. We review current knowledge regarding the roles of chaperones such as heat shock protein 90 (Hsp90) and Hsp70 and their cochaperones in cancer. Cochaperones are potential targets for cancer therapy in themselves and can be used to assess the likely prognosis of individual malignancies. Hsp70 cochaperones Bag1, Bag3, and Hop play significant roles in the etiology of some cancers as do Hsp90 cochaperones Aha1, p23, Cdc37, and FKBP1. Others such as the J domain protein family, HspBP1, TTC4, and FKBPL appear to be associated with more benign tumor phenotypes. The key importance of cochaperones for many pathways of protein folding in cancer suggests high promise for the future development of novel pharmaceutical agents.
Collapse
Affiliation(s)
- Stuart K. Calderwood
- Division of Molecular and Cellular Biology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, 99 Brookline Avenue, Boston, MA 02215, USA
- *Stuart K. Calderwood:
| |
Collapse
|