1
|
Guerra LHA, da Costa Castro NF, Jubilato FC, Marques LA, Leonel ECR, Junior SB, Campos SGP, Rahal P, Taboga SR, Calmon MF, Vilamaior PSL. Coconut Oil Mitigates the Effects of Aging on the Mongolian Gerbil Prostate. Prostate 2025; 85:395-406. [PMID: 39718211 DOI: 10.1002/pros.24842] [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: 09/02/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is a disease linked to the hormonal imbalance that occurs during aging and over the last decades, complementary and alternative medicines have come on the scene as a treatment option for BPH, such as herbal medicines. Coconut oil has been shown to be capable of interfering in testosterone-induced BPH. However, until now there is no study of the effect of coconut oil during aging. The present study evaluated the effect of the intake of coconut oil on the prostate of aging gerbils (Meriones unguiculatus). METHODS Two experimental groups were assigned: Gavage control (GC-animals subjected to gavage with water for 1 year, n = 11) and coconut oil (CO-animals subjected to gavage with coconut oil for 1 year, n = 11). Testosterone, and estradiol serum levels were determined by ELISA assay and histopathological analysis employed Hematoxylin-Eosin. Cell proliferation index was determined by PHH3 immunohistochemistry and TUNEL assay and receptors of androgen (AR) and estrogen (ERα and ERβ) were evaluated on the prostate. RESULTS The CO group exhibited a lower prostate weight (↓16.62%), decreased thickness of the prostate muscle stroma (↓18.27%), reduced expression of both AR (↓51.32) and ERα (↓14.26%) and reduced the percentage of BPH (↓1.53%) and intraepithelial neoplasms in the prostate (↓14.24%). Coconut oil intake mitigated age-related changes and increased the rate of apoptosis in prostatic cells (↑54.32). CONCLUSIONS Coconut oil treatment throughout aging helped counteract the negative effects of aging on prostate health.
Collapse
Affiliation(s)
- Luiz Henrique Alves Guerra
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Nayara Fernanda da Costa Castro
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Fernanda Costa Jubilato
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Letícia Aparecida Marques
- Department of Chemistry and Molecular Physics, São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Ellen Cristina Rivas Leonel
- Animal Molecular and Celular Biology Unit, Ecole de médecine vétérinaire, Faculté des Sciences, Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Stanislau Bogusz Junior
- Department of Chemistry and Molecular Physics, São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Silvana Gisele Pegorin Campos
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Paula Rahal
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Marilia Freitas Calmon
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Patrícia Simone Leite Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
2
|
Tahsin S, Sane NS, Cernyar B, Jiang L, Zohar Y, Lee BR, Miranti CK. AR loss in prostate cancer stroma mediated by NF-κB and p38-MAPK signaling disrupts stromal morphogen production. Oncogene 2024; 43:2092-2103. [PMID: 38769192 DOI: 10.1038/s41388-024-03064-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Androgen Receptor (AR) activity in prostate stroma is required to maintain prostate homeostasis. This is mediated through androgen-dependent induction and secretion of morphogenic factors that drive epithelial cell differentiation. However, stromal AR expression is lost in aggressive prostate cancer. The mechanisms leading to stromal AR loss and morphogen production are unknown. We identified TGFβ1 and TNFα as tumor-secreted factors capable of suppressing AR mRNA and protein expression in prostate stromal fibroblasts. Pharmacological and RNAi approaches identified NF-κB as the major signaling pathway involved in suppressing AR expression by TNFα. In addition, p38α- and p38δ-MAPK were identified as suppressors of AR expression independent of TNFα. Two regions of the AR promoter were responsible for AR suppression through TNFα. FGF10 and Wnt16 were identified as androgen-induced morphogens, whose expression was lost upon TNFα treatment and enhanced upon p38-MAPK inhibition. Wnt16, through non-canonical Jnk signaling, was required for prostate basal epithelial cell survival. These findings indicate that stromal AR loss is mediated by secreted factors within the TME. We identified TNFα/TGFβ as two possible factors, with TNFα mediating its effects through NF-κB or p38-MAPK to suppress AR mRNA transcription. This leads to loss of androgen-regulated stromal morphogens necessary to maintain normal epithelial homeostasis.
Collapse
Affiliation(s)
- Shekha Tahsin
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
| | - Neha S Sane
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Brent Cernyar
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, USA
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Benjamin R Lee
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
- Department of Urology, University of Arizona, Tucson, AZ, USA
| | - Cindy K Miranti
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA.
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
3
|
Zhang Y, Fan A, Li Y, Liu Z, Yu L, Guo J, Hou J, Li X, Chen W. Single-cell RNA sequencing reveals that HSD17B2 in cancer-associated fibroblasts promotes the development and progression of castration-resistant prostate cancer. Cancer Lett 2023; 566:216244. [PMID: 37244445 DOI: 10.1016/j.canlet.2023.216244] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
Castration-resistant prostate cancer (CRPC) responds poorly to existing therapy and appears as the lethal consequence of prostate cancer (PCa) progression. The tumour microenvironment (TME) has been thought to play a crucial role in CRPC progression. Here, we conducted single-cell RNA sequencing analysis on two CRPC and two hormone-sensitive prostate cancer (HSPC) samples to reveal potential leading roles in castration resistance. We described the single-cell transcriptional landscape of PCa. Higher cancer heterogeneity was explored in CRPC, with stronger cell cycling status and heavier copy number variant burden of luminal cells. Cancer-associated fibroblasts (CAFs), which are one of the most critical components of TME, demonstrated unique expression and cell-cell communication features in CRPC. A CAFs subtype with high expression of HSD17B2 in CRPC was identified with inflammatory features. HSD17B2 catalyses the conversion of testosterone and dihydrotestosterone to their less active forms, which was associated with steroid hormone metabolism in PCa tumour cells. However, the characteristics of HSD17B2 in PCa fibroblasts remained unknown. We found that HSD17B2 knockdown in CRPC-CAFs could inhibit migration, invasion, and castration resistance of PCa cells in vitro. Further study showed that HSD17B2 could regulate CAFs functions and promote PCa migration through the AR/ITGBL1 axis. Overall, our study revealed the important role of CAFs in the formation of CRPC. HSD17B2 in CAFs regulated AR activation and subsequent ITGBL1 secretion to promote the malignant behaviour of PCa cells. HSD17B2 in CAFs could serve as a promising therapeutic target for CRPC.
Collapse
Affiliation(s)
- Yunyan Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Aoyu Fan
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunpeng Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhuolin Liu
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liu Yu
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Hou
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaobo Li
- School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Wei Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Gangavarapu KJ, Jowdy PF, Foster BA, Huss WJ. Role of prostate stem cells and treatment strategies in benign prostate hyperplasia. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:154-169. [PMID: 35874288 PMCID: PMC9301063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/25/2022] [Indexed: 06/15/2023]
Abstract
Benign prostate hyperplasia (BPH) is a progressive disease with a direct correlation between incidence and age. Since the treatment and management of BPH involve harmful side effects and decreased quality of life for the patient, the primary focus of research should be to find better and longer-lasting therapeutic options. The mechanisms regulating prostate stem cells in development can be exploited to decrease prostate growth. BPH is defined as the overgrowth of the prostate, and BPH is often diagnosed when lower urinary tract symptoms (LUTS) of urine storage or voiding symptoms cause patients to seek treatment. While multiple factors are involved in the hyperplastic growth of the stromal and epithelial compartments of the prostate, the clonal proliferation of stem cells is considered one of the main reasons for BPH initiation and regrowth of the prostate after therapies for BPH fail. Several theories explain possible reasons for the involvement of stem cells in the development, progression, and pathogenesis of BPH. The aim of the current review is to discuss current literature on the fundamentals of prostate development and the role of stem cells in BPH. This review examines the rationale for the hypothesis that unregulated stem cell properties can lead to BPH and therapeutic targeting of stem cells may reduce treatment-related side effects and prevent the regrowth of the prostate.
Collapse
Affiliation(s)
- Kalyan J Gangavarapu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
| | - Peter F Jowdy
- Department of Dermatology, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
- Jacobs School of Medicine and Biomedical Sciences, University at BuffaloBuffalo, NY 14203, USA
| | - Barbara A Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
| | - Wendy J Huss
- Department of Dermatology, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
| |
Collapse
|
5
|
Zhang Y, Andrade R, Hanna AA, Pflum MKH. Evidence that HDAC7 acts as an epigenetic "reader" of AR acetylation through NCoR-HDAC3 dissociation. Cell Chem Biol 2022; 29:1162-1173.e5. [PMID: 35709754 DOI: 10.1016/j.chembiol.2022.05.008] [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: 03/31/2020] [Revised: 11/30/2021] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Histone deacetylase (HDAC) proteins are epigenetic regulators that govern a wide variety of cellular events. With a role in cancer formation, HDAC inhibitors have emerged as anti-cancer therapeutics. Among the eleven metal-dependent class I, II, and IV HDAC proteins targeted by inhibitor drugs, class IIa HDAC4, -5, -7, and -9 harbor low deacetylase activity and are hypothesized to be "reader" proteins, which bind to post-translationally acetylated lysine. However, evidence linking acetyllysine binding to a downstream functional event is lacking. Here, we report for the first time that HDAC4, -5, and -7 dissociated from corepressor NCoR in the presence of an acetyllysine-containing peptide, consistent with reader function. Documenting the biological consequences of this possible reader function, mutation of a critical acetylation site regulated androgen receptor (AR) transcriptional activation function through HDAC7-NCoR-HDAC3 dissociation. The data document the first evidence consistent with epigenetic-reader functions of class IIa HDAC proteins.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Rafael Andrade
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Anthony A Hanna
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA.
| |
Collapse
|
6
|
Buskin A, Singh P, Lorenz O, Robson C, Strand DW, Heer R. A Review of Prostate Organogenesis and a Role for iPSC-Derived Prostate Organoids to Study Prostate Development and Disease. Int J Mol Sci 2021; 22:ijms222313097. [PMID: 34884905 PMCID: PMC8658468 DOI: 10.3390/ijms222313097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
The prostate is vulnerable to two major age-associated diseases, cancer and benign enlargement, which account for significant morbidity and mortality for men across the globe. Prostate cancer is the most common cancer reported in men, with over 1.2 million new cases diagnosed and 350,000 deaths recorded annually worldwide. Benign prostatic hyperplasia (BPH), characterised by the continuous enlargement of the adult prostate, symptomatically afflicts around 50% of men worldwide. A better understanding of the biological processes underpinning these diseases is needed to generate new treatment approaches. Developmental studies of the prostate have shed some light on the processes essential for prostate organogenesis, with many of these up- or downregulated genes expressions also observed in prostate cancer and/or BPH progression. These insights into human disease have been inferred through comparative biological studies relying primarily on rodent models. However, directly observing mechanisms of human prostate development has been more challenging due to limitations in accessing human foetal material. Induced pluripotent stem cells (iPSCs) could provide a suitable alternative as they can mimic embryonic cells, and iPSC-derived prostate organoids present a significant opportunity to study early human prostate developmental processes. In this review, we discuss the current understanding of prostate development and its relevance to prostate-associated diseases. Additionally, we detail the potential of iPSC-derived prostate organoids for studying human prostate development and disease.
Collapse
Affiliation(s)
- Adriana Buskin
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman Building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.S.); (C.R.)
- Correspondence: (A.B.); (R.H.)
| | - Parmveer Singh
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman Building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.S.); (C.R.)
| | - Oliver Lorenz
- Newcastle University School of Computing, Digital Institute, Urban Sciences Building, Newcastle University, Newcastle upon Tyne NE4 5TG, UK;
| | - Craig Robson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman Building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.S.); (C.R.)
| | - Douglas W. Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O’Gorman Building, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.S.); (C.R.)
- Department of Urology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
- Correspondence: (A.B.); (R.H.)
| |
Collapse
|
7
|
de Almeida A, Parthimos D, Dew H, Smart O, Wiltshire M, Errington RJ. Aquaglyceroporin-3's Expression and Cellular Localization Is Differentially Modulated by Hypoxia in Prostate Cancer Cell Lines. Cells 2021; 10:cells10040838. [PMID: 33917751 PMCID: PMC8068192 DOI: 10.3390/cells10040838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/31/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
Aquaporins are required by cells to enable fast adaptation to volume and osmotic changes, as well as microenvironmental metabolic stimuli. Aquaglyceroporins play a crucial role in supplying cancer cells with glycerol for metabolic needs. Here, we show that AQP3 is differentially expressed in cells of a prostate cancer panel. AQP3 is located at the cell membrane and cytoplasm of LNCaP cell while being exclusively expressed in the cytoplasm of Du145 and PC3 cells. LNCaP cells show enhanced hypoxia growth; Du145 and PC3 cells display stress factors, indicating a crucial role for AQP3 at the plasma membrane in adaptation to hypoxia. Hypoxia, both acute and chronic affected AQP3′s cellular localization. These outcomes were validated using a machine learning classification approach of the three cell lines and of the six normoxic or hypoxic conditions. Classifiers trained on morphological features derived from cytoskeletal and nuclear labeling alongside corresponding texture features could uniquely identify each individual cell line and the corresponding hypoxia exposure. Cytoskeletal features were 70–90% accurate, while nuclear features allowed for 55–70% accuracy. Cellular texture features (73.9% accuracy) were a stronger predictor of the hypoxic load than the AQP3 distribution (60.3%).
Collapse
|
8
|
Current Status and Future Perspectives of Androgen Receptor Inhibition Therapy for Prostate Cancer: A Comprehensive Review. Biomolecules 2021; 11:biom11040492. [PMID: 33805919 PMCID: PMC8064397 DOI: 10.3390/biom11040492] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The androgen receptor (AR) is one of the main components in the development and progression of prostate cancer (PCa), and treatment strategies are mostly directed toward manipulation of the AR pathway. In the metastatic setting, androgen deprivation therapy (ADT) is the foundation of treatment in patients with hormone-sensitive prostate cancer (HSPC). However, treatment response is short-lived, and the majority of patients ultimately progress to castration-resistant prostate cancer (CRPC). Surmountable data from clinical trials have shown that the maintenance of AR signaling in the castration environment is accountable for disease progression. Study results indicate multiple factors and survival pathways involved in PCa. Based on these findings, the alternative molecular pathways involved in PCa progression can be manipulated to improve current regimens and develop novel treatment modalities in the management of CRPC. In this review, the interaction between AR signaling and other molecular pathways involved in tumor pathogenesis and its clinical implications in metastasis and advanced disease will be discussed, along with a thorough overview of current and ongoing novel treatments for AR signaling inhibition.
Collapse
|
9
|
Linares J, Marín-Jiménez JA, Badia-Ramentol J, Calon A. Determinants and Functions of CAFs Secretome During Cancer Progression and Therapy. Front Cell Dev Biol 2021; 8:621070. [PMID: 33553157 PMCID: PMC7862334 DOI: 10.3389/fcell.2020.621070] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple lines of evidence are indicating that cancer development and malignant progression are not exclusively epithelial cancer cell-autonomous processes but may also depend on crosstalk with the surrounding tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are abundantly represented in the TME and are continuously interacting with cancer cells. CAFs are regulating key mechanisms during progression to metastasis and response to treatment by enhancing cancer cells survival and aggressiveness. The latest advances in CAFs biology are pointing to CAFs-secreted factors as druggable targets and companion tools for cancer diagnosis and prognosis. Especially, extensive research conducted in the recent years has underscored the potential of several cytokines as actionable biomarkers that are currently evaluated in the clinical setting. In this review, we explore the current understanding of CAFs secretome determinants and functions to discuss their clinical implication in oncology.
Collapse
Affiliation(s)
- Jenniffer Linares
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Juan A. Marín-Jiménez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO) - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Badia-Ramentol
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
10
|
Özdemir BC. Androgen Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:169-183. [PMID: 33123999 DOI: 10.1007/978-3-030-47189-7_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The key function of mesenchymal/stromal androgen receptor (AR) signaling for prostate development has been well documented by tissue recombination experiments. Some studies have addressed the expression and function of AR in stromal cells in prostate cancer, yet our understanding of the role of stromal AR in other tissues beyond prostate is still insufficient.Genomic analysis has revealed that cellular responses to androgens differ between epithelial and stromal cells. AR in stromal cells seems not to act via classical AR transcription factors such as FOXA1 but rather depends on the JUN/AP1 complex. Stromal AR appears to have tumor-promoting and tumor-protective functions depending on tumor stage. Loss of AR signaling in fibroblasts has been detected already in premalignant lesions in the skin and prostate and has been associated with tumor induction in xenografts of skin cancer and aggressive disease features and poor patient prognosis in prostate cancer. Moreover, AR expression is found on virtually all tissue-infiltrating immune cells and plays critical roles in immune cell function. These findings suggest a potential deleterious impact of current androgen deprivation therapies which inhibit both epithelial and stromal AR, highlighting the need to develop tissue-specific AR inhibitors.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. .,International Cancer Prevention Institute, Epalinges, Switzerland.
| |
Collapse
|
11
|
Aurilio G, Cimadamore A, Mazzucchelli R, Lopez-Beltran A, Verri E, Scarpelli M, Massari F, Cheng L, Santoni M, Montironi R. Androgen Receptor Signaling Pathway in Prostate Cancer: From Genetics to Clinical Applications. Cells 2020; 9:E2653. [PMID: 33321757 PMCID: PMC7763510 DOI: 10.3390/cells9122653] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 01/10/2023] Open
Abstract
Around 80-90% of prostate cancer (PCa) cases are dependent on androgens at initial diagnosis; hence, androgen ablation therapy directed toward a reduction in serum androgens and the inhibition of androgen receptor (AR) is generally the first therapy adopted. However, the patient's response to androgen ablation therapy is variable, and 20-30% of PCa cases become castration resistant (CRPCa). Several mechanisms can guide treatment resistance to anti-AR molecules. In this regard, AR-dependent and -independent resistance mechanisms can be distinguished within the AR pathway. In this article, we investigate the multitude of AR signaling aspects, encompassing the biological structure of AR, current AR-targeted therapies, mechanisms driving resistance to AR, and AR crosstalk with other pathways, in an attempt to provide a comprehensive review for the PCa research community. We also summarize the new anti-AR drugs approved in non-metastatic castration-resistant PCa, in the castration-sensitive setting, and combination therapies with other drugs.
Collapse
Affiliation(s)
- Gaetano Aurilio
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.A.); (E.V.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (R.M.); (M.S.)
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (R.M.); (M.S.)
| | | | - Elena Verri
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.A.); (E.V.)
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (R.M.); (M.S.)
| | - Francesco Massari
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (R.M.); (M.S.)
| |
Collapse
|
12
|
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
|
13
|
MAOA-mediated reprogramming of stromal fibroblasts promotes prostate tumorigenesis and cancer stemness. Oncogene 2020; 39:3305-3321. [PMID: 32066880 DOI: 10.1038/s41388-020-1217-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/05/2023]
Abstract
The tumor microenvironment plays a critical role in prostate cancer (PC) development and progression. Inappropriate activation of the stroma potentiates the growth and transformation of epithelial tumor cells. Here, we show that upregulation of monoamine oxidase A (MAOA), a mitochondrial enzyme that degrades monoamine neurotransmitters and dietary amines, in stromal cells elevates production of reactive oxygen species, triggers an inflammatory response including activation of IL-6, and promotes tumorigenesis in vitro and in vivo. Mechanistically, MAOA enhances IL-6 transcription through direct Twist1 binding to a conserved E-box element at the IL-6 promoter. MAOA in stromal fibroblasts provides tumor cell growth advantages through paracrine IL-6/STAT3 signaling. Tissue microarray analysis revealed co-expression correlations between individual pairs of proteins of the stromal MAOA-induced Twist1/IL-6/STAT3 pathway in clinical specimens. Downstream of stromal MAOA, STAT3 also promotes cell stemness and transcriptionally activates expression of cancer stem cell marker CD44 in PC cells. MAOA inhibitor treatment effectively suppressed prostate tumor growth in mice in a stroma-specific targeted manner. Collectively, these findings characterize the contribution of MAOA to stromal activation in PC pathogenesis and provide a rationale for targeting MAOA in stromal cells to treat PC.
Collapse
|
14
|
Harris KL, Myers MB, McKim KL, Elespuru RK, Parsons BL. Rationale and Roadmap for Developing Panels of Hotspot Cancer Driver Gene Mutations as Biomarkers of Cancer Risk. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:152-175. [PMID: 31469467 PMCID: PMC6973253 DOI: 10.1002/em.22326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 05/24/2023]
Abstract
Cancer driver mutations (CDMs) are necessary and causal for carcinogenesis and have advantages as reporters of carcinogenic risk. However, little progress has been made toward developing measurements of CDMs as biomarkers for use in cancer risk assessment. Impediments for using a CDM-based metric to inform cancer risk include the complexity and stochastic nature of carcinogenesis, technical difficulty in quantifying low-frequency CDMs, and lack of established relationships between cancer driver mutant fractions and tumor incidence. Through literature review and database analyses, this review identifies the most promising targets to investigate as biomarkers of cancer risk. Mutational hotspots were discerned within the 20 most mutated genes across the 10 deadliest cancers. Forty genes were identified that encompass 108 mutational hotspot codons overrepresented in the COSMIC database; 424 different mutations within these hotspot codons account for approximately 63,000 tumors and their prevalence across tumor types is described. The review summarizes literature on the prevalence of CDMs in normal tissues and suggests such mutations are direct and indirect substrates for chemical carcinogenesis, which occurs in a spatially stochastic manner. Evidence that hotspot CDMs (hCDMs) frequently occur as tumor subpopulations is presented, indicating COSMIC data may underestimate mutation prevalence. Analyses of online databases show that genes containing hCDMs are enriched in functions related to intercellular communication. In its totality, the review provides a roadmap for the development of tissue-specific, CDM-based biomarkers of carcinogenic potential, comprised of batteries of hCDMs and can be measured by error-correct next-generation sequencing. Environ. Mol. Mutagen. 61:152-175, 2020. Published 2019. This article is a U.S. Government work and is in the public domain in the USA. Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Kelly L. Harris
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Meagan B. Myers
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Karen L. McKim
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Rosalie K. Elespuru
- Division of Biology, Chemistry and Materials ScienceCDRH/OSEL, US Food and Drug AdministrationSilver SpringMaryland
| | - Barbara L. Parsons
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| |
Collapse
|
15
|
Ishii K, Matsuoka I, Sasaki T, Nishikawa K, Kanda H, Imai H, Hirokawa Y, Iguchi K, Arima K, Sugimura Y. Loss of Fibroblast-Dependent Androgen Receptor Activation in Prostate Cancer Cells is Involved in the Mechanism of Acquired Resistance to Castration. J Clin Med 2019; 8:jcm8091379. [PMID: 31484364 PMCID: PMC6780155 DOI: 10.3390/jcm8091379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023] Open
Abstract
Loss of androgen receptor (AR) dependency in prostate cancer (PCa) cells is associated with progression to castration-resistant prostate cancer (CRPC). The tumor stroma is enriched in fibroblasts that secrete AR-activating factors. To investigate the roles of fibroblasts in AR activation under androgen deprivation, we used three sublines of androgen-sensitive LNCaP cells (E9 and F10 cells: low androgen sensitivity; and AIDL cells: androgen insensitivity) and original fibroblasts derived from patients with PCa. We performed in vivo experiments using three sublines of LNCaP cells and original fibroblasts to form homotypic tumors. The volume of tumors derived from E9 cells plus fibroblasts was reduced following androgen deprivation therapy (ADT), whereas that of F10 or AIDL cells plus fibroblasts was increased even after ADT. In tumors derived from E9 cells plus fibroblasts, serum prostate-specific antigen (PSA) decreased rapidly after ADT, but was still detectable. In contrast, serum PSA was increased even in F10 cells inoculated alone. In indirect cocultures with fibroblasts, PSA production was increased in E9 cells. Epidermal growth factor treatment stimulated Akt and p44/42 mitogen-activated protein kinase phosphorylation in E9 cells. Notably, AR splice variant 7 was detected in F10 cells. Overall, we found that fibroblast-secreted AR-activating factors modulated AR signaling in E9 cells after ADT and loss of fibroblast-dependent AR activation in F10 cells may be responsible for CRPC progression.
Collapse
Affiliation(s)
- Kenichiro Ishii
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Izumi Matsuoka
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Kohei Nishikawa
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Hideki Kanda
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Hiroshi Imai
- Pathology Division, Mie University Hospital, Tsu, Mie 514-8507, Japan.
| | - Yoshifumi Hirokawa
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Kazuhiro Iguchi
- Laboratory of Community Pharmacy, Gifu Pharmaceutical University, Gifu, Gifu 501-1196, Japan.
| | - Kiminobu Arima
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Yoshiki Sugimura
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
16
|
Nash C, Boufaied N, Badescu D, Wang YC, Paliouras M, Trifiro M, Ragoussis I, Thomson AA. Genome-wide analysis of androgen receptor binding and transcriptomic analysis in mesenchymal subsets during prostate development. Dis Model Mech 2019; 12:12/7/dmm039297. [PMID: 31350272 PMCID: PMC6679388 DOI: 10.1242/dmm.039297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate development is controlled by androgens, the androgen receptor (AR) and mesenchymal–epithelial signalling. We used chromatin immunoprecipitation sequencing (ChIP-seq) to define AR genomic binding in the male and female mesenchyme. Tissue- and single-cell-based transcriptional profiling was used to define mesenchymal AR target genes. We observed significant AR genomic binding in females and a strong enrichment at proximal promoters in both sexes. In males, there was greater AR binding to introns and intergenic regions as well as to classical AR binding motifs. In females, there was increased proximal promoter binding and involvement of cofactors. Comparison of AR-bound genes with transcriptomic data enabled the identification of novel sexually dimorphic AR target genes. We validated the dimorphic expression of AR target genes using published datasets and confirmed regulation by androgens using ex vivo organ cultures. AR targets showed variable expression in patients with androgen insensitivity syndrome. We examined AR function at single-cell resolution using single-cell RNA sequencing (scRNA-seq) in male and female mesenchyme. Surprisingly, both AR and target genes were distributed throughout cell subsets, with few positive cells within each subset. AR binding was weakly correlated with target gene expression. Summary: A study of how androgens lead to sexually dimorphic development of the prostate using transcription factor genome binding and transcriptome analysis in mesenchymal subsets.
Collapse
Affiliation(s)
- Claire Nash
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec, Canada H4A 3J1
| | - Nadia Boufaied
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec, Canada H4A 3J1
| | - Dunarel Badescu
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada H3A 0G1
| | - Yu Chang Wang
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada H3A 0G1
| | - Miltiadis Paliouras
- Division of Endocrinology, Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, 5750 Côte-des-Neiges Rd, Montreal, QC, Canada H3S 1Y9
| | - Mark Trifiro
- Division of Endocrinology, Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, 5750 Côte-des-Neiges Rd, Montreal, QC, Canada H3S 1Y9
| | - Ioannis Ragoussis
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada H3A 0G1
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec, Canada H4A 3J1
| |
Collapse
|
17
|
Wang T, Zhang FS, Ye HY, An LZ, Xiong LL, Huang XB, Xu QQ. Targeting silencing androgen receptor gene by shRNA with low-intensity focused ultrasonic irradiation inhibits growth of prostate cancer xenografts in nude mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1295-1304. [PMID: 31933943 PMCID: PMC6947058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/21/2019] [Indexed: 06/10/2023]
Abstract
The androgen receptor (AR) plays a pivotal role in prostate cancer, making it a potential therapeutic target. Short-hairpin RNA (shRNA) inhibits gene expression and offers a novel strategy to eradicate disease. Ultrasound-mediated gene transfection is a promising gene delivery method. This study sought to determine whether targeting silencing androgen receptor gene by shRNA with low-intensity focused ultrasonic irradiation could be used as effective therapy for prostate cancers in vivo. A plasmid-based short-hairpin RNA combined with low-intensity focused ultrasonic irradiation approach was used to specifically knock down the expression of AR in prostate cancer 22RV1 cells in vivo. The growth of 22RV1 tumors that had been subcutaneously xenografted was evaluated and expression level of AR was determined by immunohistochemical staining. The proliferative index (PI) and the apoptotic index (AI) were respectively derived from the percentage of positive cells by Ki-67 immunohistochemical staining and TUNEL assay. The plasmid-based AR shRNA administrated intravenously significantly inhibited the tumor growth and AR expression. These inhibitory effects of AR shRNA were augmented when the region of tumor received low-intensity focused ultrasound irradiation. Immunohistochemical staining and TUNEL assay confirmed AR shRNA with low-intensity focused ultrasonic irradiation exhibited growth-inhibitory, antiproliferative, and apoptotic effects on prostate cancer xenografts. The authors showed for the first time that the knockdown of AR expression by plasmid-based AR shRNA with low-intensity focused ultrasonic irradiation significantly suppressed the tumor growth of prostate cancer in vivo.
Collapse
Affiliation(s)
- Tian Wang
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Feng-Shi Zhang
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Hai-Yun Ye
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Li-Zhe An
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Liu-Lin Xiong
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Xiao-Bo Huang
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| | - Qing-Quan Xu
- Urology and Lithotripsy Center, Peking University People's Hospital Beijing, China
| |
Collapse
|
18
|
McAllister MJ, Underwood MA, Leung HY, Edwards J. A review on the interactions between the tumor microenvironment and androgen receptor signaling in prostate cancer. Transl Res 2019; 206:91-106. [PMID: 30528321 DOI: 10.1016/j.trsl.2018.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/23/2018] [Accepted: 11/19/2018] [Indexed: 12/29/2022]
Abstract
Prostate cancer growth is controlled by androgen receptor signaling via both androgen-dependent and androgen-independent pathways. Furthermore, the prostate is an immune competent organ with inflammatory changes both within the systemic and local environment contributing to the reprogramming of the prostatic epithelium with consistently elevated lymphocyte infiltration and proinflammatory cytokines being found in prostate cancer. The crosstalk between the tumor microenvironment and androgen receptor signaling is complex with both protumorigenic and antitumorigenic roles observed. However, despite an increase in immune checkpoint inhibitors and inflammatory signaling blockades available for a range of cancer types, we are yet to see substantial progress in the treatment of prostate cancer. Therefore, this review aims to summarize the tumor microenvironment and its impact on androgen receptor signaling in prostate cancer.
Collapse
Affiliation(s)
- Milly J McAllister
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | - Mark A Underwood
- Department of Urology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Hing Y Leung
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Urology, Queen Elizabeth University Hospital, Glasgow, United Kingdom; Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Joanne Edwards
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
19
|
The Importance of Time to Prostate-Specific Antigen (PSA) Nadir after Primary Androgen Deprivation Therapy in Hormone-Naïve Prostate Cancer Patients. J Clin Med 2018; 7:jcm7120565. [PMID: 30567361 PMCID: PMC6306761 DOI: 10.3390/jcm7120565] [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: 11/26/2018] [Revised: 12/08/2018] [Accepted: 12/18/2018] [Indexed: 11/18/2022] Open
Abstract
Prostate-specific antigen (PSA) is currently the most useful biomarker for detection of prostate cancer (PCa). The ability to measure serum PSA levels has affected all aspects of PCa management over the past two decades. The standard initial systemic therapy for advanced PCa is androgen-deprivation therapy (ADT). Although PCa patients with metastatic disease initially respond well to ADT, they often progress to castration-resistant prostate cancer (CRPC), which has a high mortality rate. We have demonstrated that time to PSA nadir (TTN) after primary ADT is an important early predictor of overall survival and progression-free survival for advanced PCa patients. In in vivo experiments, we demonstrated that the presence of fibroblasts in the PCa tumor microenvironment can prolong the period for serum PSA decline after ADT, and enhance the efficacy of ADT. Clarification of the mechanisms that affect TTN after ADT could be useful to guide selection of optimal PCa treatment strategies. In this review, we discuss recent in vitro and in vivo findings concerning the involvement of stromal–epithelial interactions in the biological mechanism of TTN after ADT to support the novel concept of “tumor regulating fibroblasts”.
Collapse
|
20
|
Nash C, Boufaied N, Mills IG, Franco OE, Hayward SW, Thomson AA. Genome-wide analysis of AR binding and comparison with transcript expression in primary human fetal prostate fibroblasts and cancer associated fibroblasts. Mol Cell Endocrinol 2018; 471:1-14. [PMID: 28483704 DOI: 10.1016/j.mce.2017.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/27/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
Abstract
The androgen receptor (AR) is a transcription factor, and key regulator of prostate development and cancer, which has discrete functions in stromal versus epithelial cells. AR expressed in mesenchyme is necessary and sufficient for prostate development while loss of stromal AR is predictive of prostate cancer progression. Many studies have characterized genome-wide binding of AR in prostate tumour cells but none have used primary mesenchyme or stroma. We applied ChIPseq to identify genomic AR binding sites in primary human fetal prostate fibroblasts and patient derived cancer associated fibroblasts, as well as the WPMY1 cell line overexpressing AR. We identified AR binding sites that were specific to fetal prostate fibroblasts (7534), cancer fibroblasts (629), WPMY1-AR (2561) as well as those common among all (783). Primary fibroblasts had a distinct AR binding profile versus prostate cancer cell lines and tissue, and showed a localisation to gene promoter binding sites 1 kb upstream of the transcriptional start site, as well as non-classical AR binding sequence motifs. We used RNAseq to define transcribed genes associated with AR binding sites and derived cistromes for embryonic and cancer fibroblasts as well as a cistrome common to both. These were compared to several in vivo ChIPseq and transcript expression datasets; which identified subsets of AR targets that were expressed in vivo and regulated by androgens. This analysis enabled us to deconvolute stromal AR targets active in stroma within tumour samples. Taken together, our data suggest that the AR shows significantly different genomic binding site locations in primary prostate fibroblasts compared to that observed in tumour cells. Validation of our AR binding site data with transcript expression in vitro and in vivo suggests that the AR target genes we have identified in primary fibroblasts may contribute to clinically significant and biologically important AR-regulated changes in prostate tissue.
Collapse
Affiliation(s)
- Claire Nash
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the McGill University Health Centre Research Institute, Montreal, Quebec, H4A 3J1, Canada
| | - Nadia Boufaied
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the McGill University Health Centre Research Institute, Montreal, Quebec, H4A 3J1, Canada
| | - Ian G Mills
- Movember/Prostate Cancer UK Centre of Excellence for Prostate Cancer Research, Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, 1001 University Place, Evanston, IL 60201, USA
| | - Simon W Hayward
- Department of Surgery, NorthShore University HealthSystem Research Institute, 1001 University Place, Evanston, IL 60201, USA
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the McGill University Health Centre Research Institute, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
21
|
Cioni B, Zwart W, Bergman AM. Androgen receptor moonlighting in the prostate cancer microenvironment. Endocr Relat Cancer 2018; 25:R331-R349. [PMID: 29618577 DOI: 10.1530/erc-18-0042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023]
Abstract
Androgen receptor (AR) signaling is vital for the normal development of the prostate and is critically involved in prostate cancer (PCa). AR is not only found in epithelial prostate cells but is also expressed in various cells in the PCa-associated stroma, which constitute the tumor microenvironment (TME). In the TME, AR is expressed in fibroblasts, macrophages, lymphocytes and neutrophils. AR expression in the TME was shown to be decreased in higher-grade and metastatic PCa, suggesting that stromal AR plays a protective role against PCa progression. With that, the functionality of AR in stromal cells appears to deviate from the receptor's classical function as described in PCa cells. However, the biological action of AR in these cells and its effect on cancer progression remains to be fully understood. Here, we systematically review the pathological, genomic and biological literature on AR actions in various subsets of prostate stromal cells and aim to better understand the consequences of AR signaling in the TME in relation to PCa development and progression.
Collapse
Affiliation(s)
- B Cioni
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - W Zwart
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode InstituteThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A M Bergman
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Regulation of masculinization: androgen signalling for external genitalia development. Nat Rev Urol 2018; 15:358-368. [DOI: 10.1038/s41585-018-0008-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Palethorpe HM, Leach DA, Need EF, Drew PA, Smith E. Myofibroblast androgen receptor expression determines cell survival in co-cultures of myofibroblasts and prostate cancer cells in vitro. Oncotarget 2018; 9:19100-19114. [PMID: 29721186 PMCID: PMC5922380 DOI: 10.18632/oncotarget.24913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 01/18/2023] Open
Abstract
Fibroblasts express androgen receptor (AR) in the normal prostate and during prostate cancer development. We have reported that loss of AR expression in prostate cancer-associated fibroblasts is a poor prognostic indicator. Here we report outcomes of direct and indirect co-cultures of immortalised AR-positive (PShTert-AR) or AR-negative (PShTert) myofibroblasts with prostate cancer cells. In the initial co-cultures the AR-negative PC3 cell line was used so AR expression and signalling were restricted to the myofibroblasts. In both direct and indirect co-culture with PShTert-AR myofibroblasts, paracrine signalling to the PC3 cells slowed proliferation and induced apoptosis. In contrast, PC3 cells proliferated with PShTert myofibroblasts irrespective of the co-culture method. In direct co-culture PC3 cells induced apoptosis in and destroyed PShTerts by direct signalling. Similar results were seen in direct co-cultures with AR-negative DU145 and AR-positive LNCaP and C4-2B prostate cancer cell lines. The AR ligand 5α-dihydrotestosterone (DHT) inhibited the proliferation of the PShTert-AR myofibroblasts, thereby reducing the extent of their inhibitory effect on cancer cell growth. These results suggest loss of stromal AR would favour prostate cancer cell growth in vivo, providing an explanation for the clinical observation that reduced stromal AR is associated with a poorer outcome.
Collapse
Affiliation(s)
- Helen M Palethorpe
- Discipline of Surgical Specialities, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Damien A Leach
- Discipline of Surgical Specialities, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia.,Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Eleanor F Need
- Discipline of Surgical Specialities, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| | - Paul A Drew
- Discipline of Surgical Specialities, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia.,School of Nursing and Midwifery, Flinders University, Adelaide, Australia
| | - Eric Smith
- Discipline of Surgical Specialities, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia.,Molecular Oncology, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville South, Australia
| |
Collapse
|
24
|
Ishii K, Takahashi S, Sugimura Y, Watanabe M. Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate. J Clin Med 2018; 7:jcm7040068. [PMID: 29614830 PMCID: PMC5920442 DOI: 10.3390/jcm7040068] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022] Open
Abstract
Androgens are essential for the development, differentiation, growth, and function of the prostate through epithelial–stromal interactions. However, androgen concentrations in the hypertrophic human prostate decrease significantly with age, suggesting an inverse correlation between androgen levels and proliferative diseases of the aging prostate. In elderly males, age- and/or androgen-related stromal remodeling is spontaneously induced, i.e., increased fibroblast and myofibroblast numbers, but decreased smooth muscle cell numbers in the prostatic stroma. These fibroblasts produce not only growth factors, cytokines, and extracellular matrix proteins, but also microRNAs as stromal paracrine signals that stimulate prostate epithelial cell proliferation. Surgical or chemical castration is the standard systemic therapy for patients with advanced prostate cancer. Androgen deprivation therapy induces temporary remission, but the majority of patients eventually progress to castration-resistant prostate cancer, which is associated with a high mortality rate. Androgen deprivation therapy-induced stromal remodeling may be involved in the development and progression of castration-resistant prostate cancer. In the tumor microenvironment, activated fibroblasts stimulating prostate cancer cell proliferation are called carcinoma-associated fibroblasts. In this review, we summarize the role of stromal paracrine signals in proliferative diseases of the aging human prostate and discuss the potential clinical applications of carcinoma-associated fibroblast-derived exosomal microRNAs as promising biomarkers.
Collapse
Affiliation(s)
- Kenichiro Ishii
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Sanai Takahashi
- Laboratory for Medical Engineering, Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa 240-8501, Japan.
| | - Yoshiki Sugimura
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
- Laboratory for Medical Engineering, Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa 240-8501, Japan.
| |
Collapse
|
25
|
Tyekucheva S, Bowden M, Bango C, Giunchi F, Huang Y, Zhou C, Bondi A, Lis R, Van Hemelrijck M, Andrén O, Andersson SO, Watson RW, Pennington S, Finn SP, Martin NE, Stampfer MJ, Parmigiani G, Penney KL, Fiorentino M, Mucci LA, Loda M. Stromal and epithelial transcriptional map of initiation progression and metastatic potential of human prostate cancer. Nat Commun 2017; 8:420. [PMID: 28871082 PMCID: PMC5583238 DOI: 10.1038/s41467-017-00460-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/29/2017] [Indexed: 01/02/2023] Open
Abstract
While progression from normal prostatic epithelium to invasive cancer is driven by molecular alterations, tumor cells and cells in the cancer microenvironment are co-dependent and co-evolve. Few human studies to date have focused on stroma. Here, we performed gene expression profiling of laser capture microdissected normal non-neoplastic prostate epithelial tissue and compared it to non-transformed and neoplastic low-grade and high-grade prostate epithelial tissue from radical prostatectomies, each with its immediately surrounding stroma. Whereas benign epithelium in prostates with and without tumor were similar in gene expression space, stroma away from tumor was significantly different from that in prostates without cancer. A stromal gene signature reflecting bone remodeling and immune-related pathways was upregulated in high compared to low-Gleason grade cases. In validation data, the signature discriminated cases that developed metastasis from those that did not. These data suggest that the microenvironment may influence prostate cancer initiation, maintenance, and metastatic progression.Stromal cells contribute to tumor development but the mechanisms regulating this process are still unclear. Here the authors analyze gene expression profiles in the prostate and show that stromal gene signature changes ahead of the epithelial gene signature as prostate cancer initiates and progresses.
Collapse
Affiliation(s)
- Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Clyde Bango
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Francesca Giunchi
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Chensheng Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Arrigo Bondi
- Department of Surgical Pathology, Maggiore Hospital, Largo Nigrisoli 2, 40133, Bologna, Italy
| | - Rosina Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Mieke Van Hemelrijck
- King's College London, Division of Cancer Studies, Translational Oncology & Urology Research, Guy's Hospital, London, SE1 9RT, UK
| | - Ove Andrén
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - Sven-Olof Andersson
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - R William Watson
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen Pennington
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Neil E Martin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Giovanni Parmigiani
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michelangelo Fiorentino
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Lorelei A Mucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
- The Broad Institute, 415 Main St, Cambridge, MA, 02142, USA.
| |
Collapse
|
26
|
Is There a Role for Preoperative 5 Alpha Reductase Inhibitors in Reducing Prostate Vascularity and Blood Loss? Curr Urol Rep 2017; 18:75. [PMID: 28766115 DOI: 10.1007/s11934-017-0730-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Benign prostatic hyperplasia (BPH) and the related medical problems are a major burden as health care costs and as a cause of patient morbidity. The introduction of medical therapy largely offered an alternative to surgical therapy, and these medications have been linked with multiple positive BPH-related outcomes. With ubiquitous use, however, a variety of adverse side effects and unsupported claims to these medications have been reported both in scientific literature and popular press. The use of 5 alpha reductase inhibitors (5ARIs) to reduce recurrent bleeding due to BPH is a reasonable option for men with recurrent trips to the physician or hospital. After a largely anecdotal report of their use in the preoperative period to reduce bleeding during BPH surgery, there was interest in the use of 5ARIs for this indication considering the effusive bleeding that can occur during BPH-related surgery, a dreaded and not uncommon complication. While the pathophysiology for the use of 5ARI to reduce BPH-related bleeding is sound, the actual clinical outcomes still require scrutiny to determine if the efficacy is both scientifically valid and clinically significant. This report will review the current literature on this topic and make attempts to determine if the use of a 5ARI before BPH-related surgery should be encouraged.
Collapse
|
27
|
Liao CP, Chen LY, Luethy A, Kim Y, Kani K, MacLeod AR, Gross ME. Androgen receptor in cancer-associated fibroblasts influences stemness in cancer cells. Endocr Relat Cancer 2017; 24:157-170. [PMID: 28264911 PMCID: PMC5453797 DOI: 10.1530/erc-16-0138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 02/06/2017] [Indexed: 12/16/2022]
Abstract
Androgen receptor (AR) regulation pathways are essential for supporting the growth and survival of prostate cancer cells. Recently, sub-populations of prostate cancer cells have been identified with stem cell features and are associated with the emergence of treatment-resistant prostate cancer. Here, we explored the function of AR in prostate cancer-associated fibroblasts (CAFs) relative to growth and stem cell-associated characteristics. CAFs were isolated from the murine cPten-/-L prostate cancer model and cultured with human prostate cancer epithelial (hPCa) cells. A murine-specific AR antisense oligonucleotide (ASO) was used to suppress the expression of AR in the CAF cells. CAFs express low, but significant levels of AR relative to fibroblasts derived from non-malignant tissue. CAFs promoted growth and colony formation of hPCa cells, which was attenuated by the suppression of AR expression. Surprisingly, AR-depleted CAFs promoted increased stem cell marker expression in hPCa cells. Interferon gamma (IFN-γ) and macrophage colony-stimulating factor (M-CSF) were increased in AR-depleted CAF cells and exhibited similar effects on stem cell marker expression as seen in the CAF co-culture systems. Clinically, elevated IFN-γ expression was found to correlate with histologic grade in primary prostate cancer samples. In summary, AR and androgen-dependent signaling are active in CAFs and exert significant effects on prostate cancer cells. IFN-γ and M-CSF are AR-regulated factors secreted by CAF cells, which promote the expression of stem cell markers in prostate cancer epithelial cells. Understanding how CAFs and other constituents of stromal tissue react to anti-cancer therapies may provide insight into the development and progression of prostate cancer.
Collapse
Affiliation(s)
- Chun-Peng Liao
- Lawrence J. Ellison Institute for Transformative MedicineKeck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Leng-Ying Chen
- Lawrence J. Ellison Institute for Transformative MedicineKeck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Andrea Luethy
- Lawrence J. Ellison Institute for Transformative MedicineKeck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Youngsoo Kim
- Ionis Pharmaceuticals Inc.Carlsbad, California, USA
| | - Kian Kani
- Lawrence J. Ellison Institute for Transformative MedicineKeck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Mitchell E Gross
- Lawrence J. Ellison Institute for Transformative MedicineKeck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
28
|
Rochette A, Boufaied N, Scarlata E, Hamel L, Brimo F, Whitaker HC, Ramos-Montoya A, Neal DE, Dragomir A, Aprikian A, Chevalier S, Thomson AA. Asporin is a stromally expressed marker associated with prostate cancer progression. Br J Cancer 2017; 116:775-784. [PMID: 28152543 PMCID: PMC5355923 DOI: 10.1038/bjc.2017.15] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prostate cancer shows considerable heterogeneity in disease progression and we propose that markers expressed in tumour stroma may be reliable predictors of aggressive tumour subtypes. METHODS We have used Kaplan-Meier, univariate and multivariate analysis to correlate the expression of Asporin (ASPN) mRNA and protein with prostate cancer progression in independent cohorts. We used immunohistochemistry and H scoring to document stromal localisation of ASPN in a tissue microarray and mouse prostate cancer model, and correlated expression with reactive stroma, defined using Masson Trichrome staining. We used cell cultures of primary prostate cancer fibroblasts treated with serum-free conditioned media from prostate cancer cell lines to examine regulation of ASPN mRNA in tumour stromal cells. RESULTS We observed increased expression of ASPN mRNA in a data set derived from benign vs tumour microdissected tissue, and a correlation with biochemical recurrence using Kaplan-Meier and Cox proportional hazard analysis. ASPN protein localised to tumour stroma and elevated expression of ASPN was correlated with decreased time to biochemical recurrence, in a cohort of 326 patients with a median follow up of 9.6 years. Univariate and multivariate analysis demonstrated that ASPN was correlated with progression, as were Gleason score, and clinical stage. Additionally, ASPN expression correlated with the presence of reactive stroma, suggesting that it may be a stromal marker expressed in response to the presence of tumour cells and particularly with aggressive tumour subtypes. We observed expression of ASPN in the stroma of tumours induced by p53 inhibition in a mouse model of prostate cancer, and correlation with neuroendocrine marker expression. Finally, we demonstrated that ASPN transcript expression in normal and cancer fibroblasts was regulated by conditioned media derived from the PC3, but not LNCaP, prostate cancer cell lines. CONCLUSIONS Our results suggest that ASPN is a stromally expressed biomarker that correlates with disease progression, and is observed in reactive stroma. ASPN expression in stroma may be part of a stromal response to aggressive tumour subtypes.
Collapse
Affiliation(s)
- Annie Rochette
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Nadia Boufaied
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Eleonora Scarlata
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Lucie Hamel
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Fadi Brimo
- Department of Pathology, Division of Urology, McGill University and The McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Hayley C Whitaker
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Antonio Ramos-Montoya
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - David E Neal
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Alice Dragomir
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Armen Aprikian
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Simone Chevalier
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
29
|
Leach DA, Trotta AP, Need EF, Risbridger GP, Taylor RA, Buchanan G. The prognostic value of stromal FK506-binding protein 1 and androgen receptor in prostate cancer outcome. Prostate 2017; 77:185-195. [PMID: 27718274 DOI: 10.1002/pros.23259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 09/07/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Improving our ability to predict cancer progression and response to conservative or radical intent therapy is critical if we are to prevent under or over treatment of individual patients. Whereas the majority of solid tumors now have a range of molecular and/or immunological markers to help define prognosis and treatment options, prostate cancer still relies mainly on histological grading and clinical parameters. We have recently reported that androgen receptor (AR) expression in stroma inversely associates with prostate cancer-specific survival, and that stromal AR reduces metastasis. For this paper, we tested the hypothesis that the AR-regulated gene FKBP51 could be used as a marker of AR activity to better predict outcome. METHODS Using immunohistochemistry on a cohort of 64 patient-matched benign and malignant prostate tissues, we assessed patient outcome by FKBP51 and AR levels. Immunoblot and RT-qPCR were used to demonstrate androgen regulation of FKBP51 in primary and primary human prostatic fibroblasts and fibroblast cell-lines. RESULTS As predicted by FKBP51 level, high AR activity in cancer stroma was associated with longer median survival (1,306 days) compared with high AR alone (699 days), whereas those with low AR and/or low FKBP51 did poorly (384 and 338 days, respectively). Survival could not be predicted on the basis cancer epithelial AR levels or activity, and was not associated with immunoreactivity in patient matched benign tissues. CONCLUSION FKBP51 improves the ability of stromal AR to predict prostate cancer-specific mortality. By adding additional immunological assessment, similar to what is already in place in a number of other cancers, we could better serve patients with prostate cancer in prognosis and informed treatment choices. Prostate 77:185-195, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damien A Leach
- The Basil Hetzel Institute for Translational Health Research, and Divisions of Medicine and Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Andrew P Trotta
- The Basil Hetzel Institute for Translational Health Research, and Divisions of Medicine and Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Eleanor F Need
- The Basil Hetzel Institute for Translational Health Research, and Divisions of Medicine and Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Gail P Risbridger
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Renea A Taylor
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Victoria, Australia
| | - Grant Buchanan
- The Basil Hetzel Institute for Translational Health Research, and Divisions of Medicine and Surgery, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
30
|
Leach DA, Buchanan G. Stromal Androgen Receptor in Prostate Cancer Development and Progression. Cancers (Basel) 2017; 9:cancers9010010. [PMID: 28117763 PMCID: PMC5295781 DOI: 10.3390/cancers9010010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 01/13/2023] Open
Abstract
Prostate cancer development and progression is the result of complex interactions between epithelia cells and fibroblasts/myofibroblasts, in a series of dynamic process amenable to regulation by hormones. Whilst androgen action through the androgen receptor (AR) is a well-established component of prostate cancer biology, it has been becoming increasingly apparent that changes in AR signalling in the surrounding stroma can dramatically influence tumour cell behavior. This is reflected in the consistent finding of a strong association between stromal AR expression and patient outcomes. In this review, we explore the relationship between AR signalling in fibroblasts/myofibroblasts and prostate cancer cells in the primary site, and detail the known functions, actions, and mechanisms of fibroblast AR signaling. We conclude with an evidence-based summary of how androgen action in stroma dramatically influences disease progression.
Collapse
Affiliation(s)
- Damien A Leach
- The Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Adelaide 5011, Australia.
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | - Grant Buchanan
- The Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Adelaide 5011, Australia.
- Department of Radiation Oncology, Canberra Teaching Hospital, Canberra 2605, Australia.
| |
Collapse
|
31
|
Leach DA, Panagopoulos V, Nash C, Bevan C, Thomson AA, Selth LA, Buchanan G. Cell-lineage specificity and role of AP-1 in the prostate fibroblast androgen receptor cistrome. Mol Cell Endocrinol 2017; 439:261-272. [PMID: 27634452 DOI: 10.1016/j.mce.2016.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/27/2022]
Abstract
Androgen receptor (AR) signalling in fibroblasts is important in prostate development and carcinogenesis, and is inversely related to prostate cancer mortality. However, the molecular mechanisms of AR action in fibroblasts and other non-epithelial cell types are largely unknown. The genome-wide DNA binding profile of AR in human prostate fibroblasts was identified by chromatin immunoprecipitation sequencing (ChIP-Seq), and found to be common to other fibroblast lines but disparate from AR cistromes of prostate cancer cells and tissue. Although AR binding sites specific to fibroblasts were less well conserved evolutionarily than those shared with cancer epithelia, they were likewise correlated with androgen regulation of fibroblast gene expression. Whereas FOXA1 is the key pioneer factor of AR in cancer epithelia, our data indicated that AP-1 likely plays a more important role in the AR cistrome in fibroblasts. The specificity of AP-1 and FOXA1 to binding in these cells is demonstrated using immunoblot and immunohistochemistry. Importantly, we find the fibroblast cistrome is represented in whole tissue/in vivo ChIP-seq studies at both genomic and resulting protein levels, highlighting the importance of the stroma in whole tissue -omic studies. This is the first nuclear receptor ChIP-seq study in prostatic fibroblasts, and provides novel insight into the action of fibroblast AR in prostate cancer.
Collapse
Affiliation(s)
- Damien A Leach
- The Basil Hetzel Institute for Translational Health Research, The University of Adelaide, SA, Australia; Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Vasilios Panagopoulos
- The Basil Hetzel Institute for Translational Health Research, The University of Adelaide, SA, Australia
| | - Claire Nash
- Division of Urology, Department of Surgery, McGill University Health Centre, Montreal, Canada
| | - Charlotte Bevan
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Axel A Thomson
- Division of Urology, Department of Surgery, McGill University Health Centre, Montreal, Canada
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, Adelaide, SA, Australia; Freemasons Foundation Centre for Mens' Health, School of Medicine, The University of Adelaide, Adelaide, SA, Australia.
| | - Grant Buchanan
- The Basil Hetzel Institute for Translational Health Research, The University of Adelaide, SA, Australia.
| |
Collapse
|
32
|
Torres-Estay V, Carreño DV, Fuenzalida P, Watts A, San Francisco IF, Montecinos VP, Sotomayor PC, Ebos J, Smith GJ, Godoy AS. Androgens modulate male-derived endothelial cell homeostasis using androgen receptor-dependent and receptor-independent mechanisms. Angiogenesis 2016; 20:25-38. [PMID: 27679502 DOI: 10.1007/s10456-016-9525-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/12/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Sex-related differences in the role of androgen have been reported in cardiovascular diseases and angiogenesis. Moreover, androgen receptor (AR) has been causally involved in the homeostasis of human prostate endothelial cells. However, levels of expression, functionality and biological role of AR in male- and female-derived human endothelial cells (ECs) remain poorly characterized. The objectives of this work were (1) to characterize the functional expression of AR in male- and female-derived human umbilical vein endothelial cell (HUVEC), and (2) to specifically analyze the biological effects of DHT, and the role of AR on these effects, in male-derived HUVECs (mHUVECs). RESULTS Immunohistochemical analyses of tissue microarrays from benign human tissues confirmed expression of AR in ECs from several androgen-regulated and non-androgen-regulated human organs. Functional expression of AR was validated in vitro in male- and female-derived HUVECs using quantitative RT-PCR, immunoblotting and AR-mediated transcriptional activity assays. Our results indicated that functional expression of AR in male- and female-derived HUVECs was heterogeneous, but not sex dependent. In parallel, we analyzed in depth the biological effects of DHT, and the role of AR on these effects, on proliferation, survival and tube formation capacity in mHUVECs. Our results indicated that DHT did not affect mHUVEC survival; however, DHT stimulated mHUVEC proliferation and suppressed mHUVEC tube formation capacity. While the effect of DHT on proliferation was mediated through AR, the effect of DHT on tube formation did not depend on the presence of a functional AR, but rather depended on the ability of mHUVECs to further metabolize DHT. CONCLUSIONS (1) Heterogeneous expression of AR in male- and female-derived HUVEC could define the presence of functionally different subpopulations of ECs that may be affected differentially by androgens, which could explain, at least in part, the pleiotropic effects of androgen on vascular biology, and (2) DHT, and metabolites of DHT, generally thought to represent progressively more hydrophilic products along the path to elimination, may have differential roles in modulating the biology of human ECs through AR-dependent and AR-independent mechanisms, respectively.
Collapse
Affiliation(s)
- Verónica Torres-Estay
- Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Daniela V Carreño
- Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Patricia Fuenzalida
- Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Anica Watts
- Department of Urology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Ignacio F San Francisco
- Department of Urology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Viviana P Montecinos
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Paula C Sotomayor
- Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile
| | - John Ebos
- Department of Cancer Genetics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA.,Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Gary J Smith
- Department of Urology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Alejandro S Godoy
- Department of Physiology, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile. .,Department of Urology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
33
|
Lu GQ, Wu ZB, Chu XY, Bi ZG, Fan WX. An investigation of crosstalk between Wnt/β-catenin and transforming growth factor-β signaling in androgenetic alopecia. Medicine (Baltimore) 2016; 95:e4297. [PMID: 27472703 PMCID: PMC5265840 DOI: 10.1097/md.0000000000004297] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Wnt and transforming growth factor-β (TGF-β) signaling pathways are known to be involved in the pathogenesis of androgenetic alopecia (AGA). However, the way that Wnt and TGF-β signaling is altered in patients with AGA and whether there exists a crosstalk between them in pathogenetic process of AGA remain unclear. OBJECTIVES To investigate the expression of Wnt and TGF-β signaling and the crosstalk between these 2 signaling pathways in AGA. METHODS Fifteen male patients with AGA were recruited for our research. Fifteen scalp specimens of the balding were collected from frontal areas, and 9 nonbalding were collected from occipital areas. We analyzed the expression and activation of downstream Wnt and TGF-β signaling molecules in both balding and nonbalding hair follicles isolated from scalp specimens. Furthermore, we evaluated the activation of Wnt and TGF-β signaling after either of them was blocked with the inhibitor in balding and nonbalding dermal papilla (DP) cells. RESULTS Compared with the nonbalding counterparts, the mRNA level of Wnt10a and LEF1 was decreased. But TβRI and TβRII, and the protein expression of TGF-β1 was elevated in balding hair follicles. To investigate the crosstalk between Wnt and TGF-β signaling, we used SB431542 to inhibit the TGF-β signaling in balding DP cells and found that SB431542 significantly attenuated the phosphorylation of Smad2 and Akt. However, the mRNA level of Wnt10a, LEF1, and the nuclear translocation of β-catenin was increased. On the other hand, we suppressed the Wnt signaling by XAV939 in nonbalding DP cells, which displayed that the level of β-catenin and LEF1 was significantly inhibited; however, the level of active TGF-β1 and the phosphorylation of Smad2 and Akt were up-regulated. CONCLUSIONS These data indicate that crosstalk between Wnt/β-catenin and TGF-β signaling pathways may exist as one of the important mechanisms contributing to AGA.
Collapse
Affiliation(s)
- Gui-Qing Lu
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University
- Department of Dermatology, BenQ Medical Center, Nanjing Medical University
| | - Zhi-Bo Wu
- Department of Dermatology, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Xiao-Yan Chu
- Department of Dermatology, BenQ Medical Center, Nanjing Medical University
| | - Zhi-Gang Bi
- Department of Dermatology, BenQ Medical Center, Nanjing Medical University
| | - Wei-Xin Fan
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University
- Correspondence: Wei-Xin Fan, Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Street 300#, Nanjing, Jiangsu 210029, P.R. China (e-mail: )
| |
Collapse
|
34
|
Leach DA, Need EF, Toivanen R, Trotta AP, Palethorpe HM, Palenthorpe HM, Tamblyn DJ, Kopsaftis T, England GM, Smith E, Drew PA, Pinnock CB, Lee P, Holst J, Risbridger GP, Chopra S, DeFranco DB, Taylor RA, Buchanan G. Stromal androgen receptor regulates the composition of the microenvironment to influence prostate cancer outcome. Oncotarget 2016; 6:16135-50. [PMID: 25965833 PMCID: PMC4599261 DOI: 10.18632/oncotarget.3873] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/02/2015] [Indexed: 12/21/2022] Open
Abstract
Androgen receptor (AR) signaling in stromal cells is important in prostate cancer, yet the mechanisms underpinning stromal AR contribution to disease development and progression remain unclear. Using patient-matched benign and malignant prostate samples, we show a significant association between low AR levels in cancer associated stroma and increased prostate cancer-related death at one, three and five years post-diganosis, and in tissue recombination models with primary prostate cancer cells that low stromal AR decreases castration-induced apoptosis. AR-regulation was found to be different in primary human fibroblasts isolated from adjacent to cancerous and non-cancerous prostate epithelia, and to represent altered activation of myofibroblast pathways involved in cell cycle, adhesion, migration, and the extracellular matrix (ECM). Without AR signaling, the fibroblast-derived ECM loses the capacity to promote attachment of both myofibroblasts and cancer cells, is less able to prevent cell-matrix disruption, and is less likely to impede cancer cell invasion. AR signaling in prostate cancer stroma appears therefore to alter patient outcome by maintaining an ECM microenvironment inhibitory to cancer cell invasion. This paper provides comprehensive insight into AR signaling in the non-epithelial prostate microenvironment, and a resource from which the prognostic and therapeutic implications of stromal AR levels can be further explored.
Collapse
Affiliation(s)
- Damien A Leach
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | - Eleanor F Need
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | - Roxanne Toivanen
- Department of Anatomy and Development, Monash University, VIC, Australia
| | - Andrew P Trotta
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | - Helen M Palethorpe
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | - Helen M Palenthorpe
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | | | - Tina Kopsaftis
- Urology Unit, Repatriation General Hospital, SA, Australia
| | - Georgina M England
- Department of Surgical Pathology, SA Pathology at Flinders Medical Centre, SA, Australia
| | - Eric Smith
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| | - Paul A Drew
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia.,School of Nursing and Midwifery, Flinders University, Bedford Park, SA, Australia
| | | | - Peng Lee
- Department of Pathology and Urology, New York University, NY, USA
| | - Jeff Holst
- Origins of Cancer Laboratory, Centenary Institute, NSW, Australia.,Sydney Medical School, University of Sydney, NSW, Australia
| | - Gail P Risbridger
- Department of Anatomy and Development, Monash University, VIC, Australia
| | - Samarth Chopra
- Urology Unit, Repatriation General Hospital, SA, Australia.,Department of Urology, St Vincent's Hospital, Sydney and Garvan Institute, NSW, Australia
| | - Donald B DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Renea A Taylor
- Department of Anatomy and Development, Monash University, VIC, Australia.,Department of Physiology, Monash University, VIC, Australia
| | - Grant Buchanan
- The Basil Hetzel Institute for Translational Health Research, University of Adelaide, SA, Australia
| |
Collapse
|
35
|
Fibroblasts prolong serum prostate-specific antigen decline after androgen deprivation therapy in prostate cancer. J Transl Med 2016; 96:338-49. [PMID: 26641067 DOI: 10.1038/labinvest.2015.136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/29/2015] [Accepted: 10/08/2015] [Indexed: 11/09/2022] Open
Abstract
In patients with prostate cancer (PCa), serum prostate-specific antigen (PSA) is a useful marker for evaluating the effects of androgen deprivation therapy (ADT). Intuitively, most urologists expect that a more rapid PSA decline in response to ADT would be positively associated with extended survival. Recently, we have reported that prolonged gradual serum PSA decline after ADT is strongly associated with favorable prognosis in PCa patients, however, the mechanism remains unknown. We investigated the role of fibroblasts in serum PSA decline after ADT. We performed in vitro experiments using androgen-sensitive, androgen receptor (AR)-positive prostate epithelial cell lines (LNCaP, 22Rv1, and RWPE-1 cells), commercially available prostate stromal cells (PrSC), and primary cultures of prostate fibroblasts (pcPrFs). In LNCaP and 22Rv1 cells, PSA production was increased by co-culture with fibroblasts under androgen-deprived conditions. In an in vivo model using LNCaP cells, serum PSA declined rapidly after ADT becoming undetectable within 14 days in mice inoculated with LNCaP cells alone. In contrast, when LNCaP cells were co-inoculated with fibroblasts, serum PSA levels were still high on 14 days post ADT and did not drop to undetectable levels until 21 days post ADT. Tumor volumes and Ki67 labeling indices were not altered between days 14 and 21 post ADT in mice inoculated with LNCaP cells; however, those in mice inoculated with LNCaP cells plus fibroblasts decreased gradually. PSA protein was detected in all tumors on 21 days post ADT by immunohistochemical staining. Microvessel densities were higher on 14 days post ADT for tumors from mice inoculated with LNCaP cells plus fibroblasts as compared with LNCaP cells alone. In summary, co-inoculation of fibroblasts with LNCaP cells prolonged serum PSA decline after ADT and enhanced the efficacy of ADT. Prolonged serum PSA decline may indicate the presence of protective fibroblasts that preserve the AR dependence of PCa cells, improving treatment efficacy.
Collapse
|
36
|
Timing of androgen receptor disruption and estrogen exposure underlies a spectrum of congenital penile anomalies. Proc Natl Acad Sci U S A 2015; 112:E7194-203. [PMID: 26598695 DOI: 10.1073/pnas.1515981112] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Congenital penile anomalies (CPAs) are among the most common human birth defects. Reports of CPAs, which include hypospadias, chordee, micropenis, and ambiguous genitalia, have risen sharply in recent decades, but the causes of these malformations are rarely identified. Both genetic anomalies and environmental factors, such as antiandrogenic and estrogenic endocrine disrupting chemicals (EDCs), are suspected to cause CPAs; however, little is known about the temporal window(s) of sensitivity to EDCs, or the tissue-specific roles and downstream targets of the androgen receptor (AR) in external genitalia. Here, we show that the full spectrum of CPAs can be produced by disrupting AR at different developmental stages and in specific cell types in the mouse genital tubercle. Inactivation of AR during a narrow window of prenatal development results in hypospadias and chordee, whereas earlier disruptions cause ambiguous genitalia and later disruptions cause micropenis. The neonatal phase of penile development is controlled by the balance of AR to estrogen receptor α (ERα) activity; either inhibition of androgen or augmentation of estrogen signaling can induce micropenis. AR and ERα have opposite effects on cell division, apoptosis, and regulation of Hedgehog, fibroblast growth factor, bone morphogenetic protein, and Wnt signaling in the genital tubercle. We identify Indian hedgehog (Ihh) as a novel downstream target of AR in external genitalia and show that conditional deletion of Ihh inhibits penile masculinization. These studies reveal previously unidentified cellular and molecular mechanisms by which antiandrogenic and estrogenic signals induce penile malformations and demonstrate that the timing of endocrine disruption can determine the type of CPA.
Collapse
|
37
|
Wen S, Chang HC, Tian J, Shang Z, Niu Y, Chang C. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:293-301. [PMID: 25432062 PMCID: PMC4305176 DOI: 10.1016/j.ajpath.2014.10.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 10/13/2014] [Accepted: 10/20/2014] [Indexed: 02/05/2023]
Abstract
The prostate is an androgen-sensitive organ that needs proper androgen/androgen receptor (AR) signals for normal development. The progression of prostate diseases, including benign prostate hyperplasia (BPH) and prostate cancer (PCa), also needs proper androgen/AR signals. Tissue recombination studies report that stromal, but not epithelial, AR plays more critical roles via the mesenchymal-epithelial interactions to influence the early process of prostate development. However, in BPH and PCa, much more attention has been focused on epithelial AR roles. However, accumulating evidence indicates that stromal AR is also irreplaceable and plays critical roles in prostate disease progression. Herein, we summarize the roles of stromal AR in the development of normal prostate, BPH, and PCa, with evidence from the recent results of in vitro cell line studies, tissue recombination experiments, and AR knockout animal models. Current evidence suggests that stromal AR may play positive roles to promote BPH and PCa progression, and targeting stromal AR selectively with AR degradation enhancer, ASC-J9, may allow development of better therapies with fewer adverse effects to battle BPH and PCa.
Collapse
Affiliation(s)
- Simeng Wen
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Hong-Chiang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Jing Tian
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York; Sex Hormone Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
38
|
Abstract
Androgen receptor (AR) signaling is vital to the development and function of the prostate and is a key pathway in prostate cancer. AR is differentially expressed in the stroma and epithelium, with both paracrine and autocrine control throughout the prostate. Stromal-epithelial interactions within the prostate are commonly dependent on AR signaling and expression. Alterations in these pathways can promote tumorigenesis. AR is also expressed in normal and malignant mammary tissues. Emerging data indicate a role for AR in certain subtypes of breast cancer that has the potential to be exploited therapeutically. The aim of this review is to highlight the importance of these interactions in normal development and tumorigenesis, with a focus on the prostate and breast.
Collapse
Affiliation(s)
- Cera M Nieto
- Department of PharmacologyUniversity of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Leah C Rider
- Department of PharmacologyUniversity of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Scott D Cramer
- Department of PharmacologyUniversity of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
39
|
DeGraff DJ, Grabowska MM, Case T, Yu X, Herrick MK, Hayward W, Strand DW, Cates JM, Hayward SW, Gao N, Walter MA, Buttyan R, Yi Y, Kaestner KH, Matusik RJ. FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype. J Transl Med 2014; 94:726-39. [PMID: 24840332 PMCID: PMC4451837 DOI: 10.1038/labinvest.2014.64] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/20/2014] [Accepted: 03/26/2014] [Indexed: 01/25/2023] Open
Abstract
The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.
Collapse
Affiliation(s)
- David J. DeGraff
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | | | - Tom Case
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Xiuping Yu
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Mary K. Herrick
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - William Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Douglas W. Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Justin M. Cates
- Department of Pathology, Vanderbilt University Medical Center, Nashville TN
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark NJ
| | | | | | - Yajun Yi
- Institute for Integrative Genomics and Department of Medicine, Vanderbilt University, Nashville TN
| | | | - Robert J. Matusik
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN,Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN,Correspondence and reprint requests should be made to: Robert J. Matusik, Ph.D., William L. Bray Chair of Urologic Surgery, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232,
| |
Collapse
|
40
|
Yu Y, Lee JS, Xie N, Li E, Hurtado-Coll A, Fazli L, Cox M, Plymate S, Gleave M, Dong X. Prostate stromal cells express the progesterone receptor to control cancer cell mobility. PLoS One 2014; 9:e92714. [PMID: 24664419 PMCID: PMC3963951 DOI: 10.1371/journal.pone.0092714] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 02/24/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Reciprocal interactions between epithelium and stroma play vital roles for prostate cancer development and progression. Enhanced secretions of cytokines and growth factors by cancer associated fibroblasts in prostate tumors create a favorable microenvironment for cancer cells to grow and metastasize. Our previous work showed that the progesterone receptor (PR) was expressed specifically in prostate stromal fibroblasts and smooth muscle cells. However, the expression levels of PR and its impact to tumor microenvironment in prostate tumors are poorly understood. METHODS Immunohistochemistry assays are applied to human prostate tissue biopsies. Cell migration, invasion and proliferation assays are performed using human prostate cells. Real-time PCR and ELISA are applied to measure gene expression at molecular levels. RESULTS Immunohistochemistry assays showed that PR protein levels were decreased in cancer associated stroma when compared with paired normal prostate stroma. Using in vitro prostate stromal cell models, we showed that conditioned media collected from PR positive stromal cells inhibited prostate cancer cell migration and invasion, but had minor suppressive impacts on cancer cell proliferation. PR suppressed the secretion of stromal derived factor-1 (SDF-1) and interlukin-6 (IL-6) by stromal cells independent to PR ligands. Blocking PR expression by siRNA or supplementation of exogenous SDF-1 or IL-6 to conditioned media from PR positive stromal cells counteracted the inhibitory effects of PR to cancer cell migration and invasion. CONCLUSIONS Decreased expression of the PR in cancer associated stroma may contribute to the elevated SDF-1 and IL-6 levels in prostate tumors and enhance prostate tumor progression.
Collapse
Affiliation(s)
- Yue Yu
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Suehyun Lee
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ning Xie
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Estelle Li
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Antonio Hurtado-Coll
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Cox
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen Plymate
- Department of Medicine, University of Washington School of Medicine and VAPSHCS-GRECC, Seattle, Washington, United States of America
| | - Martin Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xuesen Dong
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Mahmoud AM, Zhu T, Parray A, Siddique HR, Yang W, Saleem M, Bosland MC. Differential effects of genistein on prostate cancer cells depend on mutational status of the androgen receptor. PLoS One 2013; 8:e78479. [PMID: 24167630 PMCID: PMC3805529 DOI: 10.1371/journal.pone.0078479] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/12/2013] [Indexed: 12/21/2022] Open
Abstract
Blocking the androgen receptor (AR) activity is the main goal of therapies for advanced prostate cancer (PCa). However, relapse with a more aggressive, hormone refractory PCa arises, which harbors restored AR activity. One mechanism of such reactivation occurs through acquisition of AR mutations that enable its activation by various steroidal and non-steroidal structures. Thus, natural and chemical compounds that contribute to inappropriate (androgen-independent) activation of the AR become an area of intensive research. Here, we demonstrate that genistein, a soy phytoestrogen binds to both the wild and the Thr877Ala (T877A) mutant types of AR competitively with androgen, nevertheless, it exerts a pleiotropic effect on PCa cell proliferation and AR activity depending on the mutational status of the AR. Genistein inhibited, in a dose-dependent way, cell proliferation and AR nuclear localization and expression in LAPC-4 cells that have wild AR. However, in LNCaP cells that express the T877A mutant AR, genistein induced a biphasic effect where physiological doses (0.5-5 µmol/L) stimulated cell growth and increased AR expression and transcriptional activity, and higher doses induced inhibitory effects. Similar biphasic results were achieved in PC-3 cells transfected with AR mutants; T877A, W741C and H874Y. These findings suggest that genistein, at physiological concentrations, potentially act as an agonist and activate the mutant AR that can be present in advanced PCa after androgen ablation therapy.
Collapse
Affiliation(s)
- Abeer M. Mahmoud
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Tian Zhu
- Center of Pharmaceutical Biotechnology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Aijaz Parray
- Section of Molecular Chemoprevention and Therapeutics, the Hormel Institute, University of Minnesota, Austin, Texas, United States of America
| | - Hifzur R. Siddique
- Section of Molecular Chemoprevention and Therapeutics, the Hormel Institute, University of Minnesota, Austin, Texas, United States of America
| | - Wancai Yang
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Mohammad Saleem
- Section of Molecular Chemoprevention and Therapeutics, the Hormel Institute, University of Minnesota, Austin, Texas, United States of America
| | - Maarten C. Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
42
|
Gravina GL, Mancini A, Ranieri G, Di Pasquale B, Marampon F, Di Clemente L, Ricevuto E, Festuccia C. Phenotypic characterization of human prostatic stromal cells in primary cultures derived from human tissue samples. Int J Oncol 2013; 42:2116-22. [PMID: 23589051 DOI: 10.3892/ijo.2013.1892] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 11/14/2012] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence has shown that the tumor microenvironment plays a crucial role in prostate cancer (PCa) development and progression. However, the mechanism(s) through which stromal cells regulate epithelial cells and the differences among prostatic stromal cells of different histological/pathological origin in PCa progression remain unclear. Therefore, it is necessary to characterize the stromal cell populations present in benign prostatic hyperplasia (BPH) and PCa. To this end, we used cultures from stromal cells obtained from BPH-derived (15 cases) and PCa-derived (30 cases) primary cultures. In culture, stromal cells are a mixture of fibroblasts, myofibroblasts (MFs) and muscle cells. Fibroblasts are characterized for the expression of vimentin, MFs for the co-expression of α-smooth muscle actin (α-SMA) and vimentin, whereas muscle cells for the expression of α-SMA and desmin. Fibroblasts were present in large amounts in the BPH- compared to the PCa-derived cultures, whereas MFs were more representative of PCa- as opposed to BPH-derived cultures. Some α-SMA-positive cells retained the expression of basal cytokeratin K14. This population was defined as myoepithelial cells and was associated with senescent cultures. The percentage of MFs was higher in high-grade compared to moderate- and low-grade PCa-derived cultures, whereas the number of myoepithelial cells was lower in high-grade compared to moderate- and low-grade PCa-derived cultures. In addition, we analyzed the expression of p75NTR, as well as the expression of matrix metalloproteinase (MMP)-2, MMP-9 and tissue inhibitors of MMPs (TIMPs). p75NTR expression was elevated in the stromal cultures derived from PCa compared to those derived from BPH and in cultures derived from cases with Gleason scores ≥7 compared to those derived from cases with Gleason scores <7, as well as in cultures with a high concentration of MFs compared to those with a high concentration of fibroblasts. MMP-2 was secreted by all primary cultures, whereas MMP-9 secretion was observed only in some PCa-derived stromal cells, when the percentage of MFs was significantly higher compared to BPH-derived cultures. TIMP1, TIMP2 and TIMP3 were secreted in elevated amounts in the BPH- compared to the PCa-derived stromal cultures, suggesting the differential regulation of extracellular matrix (ECM) degradation. When we used 22rv1 and PC3 PCa xenograft models for the isolation and characterization of murine cancer-associated fibroblasts (CAFs) we noted that the angiogenic wave was concurrent with the appearance of a reactive stroma phenotype, as determined by staining for α-SMA, vimentin, tenascin, calponin, desmin and Masson's trichrome. In conclusion, MF stromal cells from PCa participate in the progression and metastasis of PCa, modualting inflammation, angiogenesis and epithelial cancer cell proliferation.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Sampson N, Neuwirt H, Puhr M, Klocker H, Eder IE. In vitro model systems to study androgen receptor signaling in prostate cancer. Endocr Relat Cancer 2013; 20:R49-64. [PMID: 23447570 DOI: 10.1530/erc-12-0401] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Prostate cancer (PCa) is one of the most common causes of male cancer-related death in Western nations. The cellular response to androgens is mediated via the androgen receptor (AR), a ligand-inducible transcription factor whose dysregulation plays a key role during PCa development and progression following androgen deprivation therapy, the current mainstay systemic treatment for advanced PCa. Thus, a better understanding of AR signaling and new strategies to abrogate AR activity are essential for improved therapeutic intervention. Consequently, a large number of experimental cell culture models have been established to facilitate in vitro investigations into the role of AR signaling in PCa development and progression. These different model systems mimic distinct stages of this heterogeneous disease and exhibit differences with respect to AR expression/status and androgen responsiveness. Technological advances have facilitated the development of in vitro systems that more closely reflect the physiological setting, for example via the use of three-dimensional coculture to study the interaction of prostate epithelial cells with the stroma, endothelium, immune system and tissue matrix environment. This review provides an overview of the most commonly used in vitro cell models currently available to study AR signaling with particular focus on their use in addressing key questions relating to the development and progression of PCa. It is hoped that the continued development of in vitro models will provide more biologically relevant platforms for mechanistic studies, drug discovery and design ensuring a more rapid transfer of knowledge from the laboratory to the clinic.
Collapse
Affiliation(s)
- Natalie Sampson
- Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Anichstraße 35, A-6020 Innsbruck, Austria
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Abstract
Reactive stroma initiates during early prostate cancer development and coevolves with prostate cancer progression. Previous studies have defined the key markers of reactive stroma and have established that reactive stroma biology influences prostate tumorigenesis and progression. The stem/progenitor cells of origin and the mechanisms that regulate their recruitment and activation to myofibroblasts or carcinoma-associated fibroblasts are essentially unknown. Key regulatory factors have been identified, including transforming growth factor β, interleukin-8, fibroblast growth factors, connective tissue growth factor, wingless homologs-Wnts, and stromal cell-derived factor-1, among others. The biology of reactive stroma in cancer is similar to the more predictable biology of the stroma compartment during wound repair at sites where the epithelial barrier function is breached and a stromal response is generated. The coevolution of reactive stroma and the biology of how reactive stroma-carcinoma interactions regulate cancer progression and metastasis are targets for new therapeutic approaches. Such approaches are strategically designed to inhibit cancer progression by uncoupling the reactive stroma niche.
Collapse
Affiliation(s)
- David A Barron
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
46
|
Mehta V, Abler LL, Keil KP, Schmitz CT, Joshi PS, Vezina CM. Atlas of Wnt and R-spondin gene expression in the developing male mouse lower urogenital tract. Dev Dyn 2011; 240:2548-60. [PMID: 21936019 DOI: 10.1002/dvdy.22741] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2011] [Indexed: 12/24/2022] Open
Abstract
Prostate development is influenced by β-catenin signaling, but it is unclear which β-catenin activators are involved, where they are synthesized, and whether their mRNA abundance is influenced by androgens. We identified WNT/β-catenin-responsive β-galactosidase activity in the lower urogenital tract (LUT) of transgenic reporter mice, but β-galactosidase activity differed among the four mouse strains we examined. We used in situ hybridization to compare patterns of Wnts, r-spondins (Rspos, co-activators of β-catenin signaling), β-catenin-responsive mRNAs, and an androgen receptor-responsive mRNA in wild type fetal male, fetal female, and neonatal male LUT. Most Wnt and Rspo mRNAs were present in LUT during prostate development. Sexually dimorphic expression patterns were observed for WNT/β-catenin-responsive genes, and for Wnt2b, Wnt4, Wnt7a, Wnt9b, Wnt10b, Wnt11, Wnt16, and Rspo3 mRNAs. These results reveal sexual differences in WNT/β-catenin signaling in fetal LUT, supporting the idea that this pathway may be directly or indirectly responsive to androgens during prostate ductal development.
Collapse
Affiliation(s)
- Vatsal Mehta
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | | | |
Collapse
|
47
|
Siddique HR, Mishra SK, Karnes RJ, Saleem M. Lupeol, a novel androgen receptor inhibitor: implications in prostate cancer therapy. Clin Cancer Res 2011; 17:5379-91. [PMID: 21712449 DOI: 10.1158/1078-0432.ccr-11-0916] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Conventional therapies to treat prostate cancer (CaP) of androgen-dependent phenotype (ADPC) and castration-resistant phenotype (CRPC) are deficient in outcome which has necessitated a need to identify those agents that could target AR for both disease types. We provide mechanism-based evidence that lupeol (Lup-20(29)-en-3b-ol) is a potent inhibitor of androgen receptor (AR) in vitro and in vivo. EXPERIMENTAL DESIGN Normal prostate epithelial cell (RWPE-1), LAPC4 (wild functional AR/ADPC), LNCaP (mutant functional/AR/ADPC), and C4-2b (mutant functional/AR/CRPC) cells were used to test the anti-AR activity of lupeol. Cells grown under androgen-rich environment and treated with lupeol were tested for proliferation, AR transcriptional activity, AR competitive ligand binding, AR-DNA binding, and AR-ARE/target gene binding. Furthermore, in silico molecular modeling for lupeol-AR binding was done. Athymic mice bearing C4-2b and LNCaP cell-originated tumors were treated intraperitoneally with lupeol (40 mg/kg; 3 times/wk) and tumor growth and surrogate biomarkers were evaluated. To assess bioavailability, lupeol serum levels were measured. RESULTS Lupeol significantly inhibited R1881 (androgen analogue) induced (i) transcriptional activity of AR and (ii) expression of PSA. Lupeol (i) competed antagonistically with androgen for AR, (ii) blocked the binding of AR to AR-responsive genes including PSA, TIPARP, SGK, and IL-6, and (iii) inhibited the recruitment of RNA Pol II to target genes. Lupeol sensitized CRPC cells to antihormone therapy. High-performance liquid chromatography analysis showed that lupeol is bioavailable to mice. Lupeol inhibited the tumorigenicity of both ADPC and CRPC cells in animals. Serum and tumor tissues exhibited reduced PSA levels. CONCLUSION Lupeol, an effective AR inhibitor, could be developed as a potential agent to treat human CaP.
Collapse
Affiliation(s)
- Hifzur Rahman Siddique
- Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, USA
| | | | | | | |
Collapse
|