1
|
Björnebo L, Razdan S, Discacciati A, Palsdottir T, Aly M, Nordström T, Eklund M, Lundon D, Grönberg H, Tewari A, Wiklund P, Kyprianou N, Lantz A. Prostate cancer incidence and mortality in men exposed to α1-adrenergic receptor antagonists. J Natl Cancer Inst 2024; 116:1459-1465. [PMID: 38718219 PMCID: PMC11378311 DOI: 10.1093/jnci/djae108] [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: 01/15/2024] [Revised: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND α1-Adrenergic receptor antagonists are commonly used to treat benign prostatic hyperplasia. Preclinical studies suggest that they induce cell death and inhibit tumor growth. This study evaluated the risk of prostate cancer death in men using α1-adrenergic receptor antagonists. METHODS A population-based cohort study in Stockholm, Sweden (January 1, 2007, to December 31, 2019) included 451 779 men with a prostate-specific antigen test result. Study entry was 1 year after the first prostate-specific antigen test. Men were considered exposed at their second filled prescription. The primary outcome was prostate cancer mortality. Secondary outcomes were all-cause mortality and prostate cancer incidence. Cox proportional hazards regression models were used to calculate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for all outcomes. Inverse-probability weighting with marginal structural models accounted for time-dependent confounders. RESULTS Of 351 297 men in the final cohort, 39 856 (11.3%) were exposed to α1-adrenergic receptor antagonists. Median (interquartile range) follow-up for prostate cancer mortality was 8.9 (5.1-10.9) years; median (interquartile range) exposure time to α1-adrenergic receptor antagonists was 4.4 (2.0-7.6) years. There was no evidence of an association between α1-adrenergic receptor antagonist use and prostate cancer mortality, all-cause mortality, or high-grade prostate cancer. α1-Adrenergic receptor antagonist use was associated with an increased risk of prostate cancer (HR = 1.11, 95% CI = 1.06 to 1.17) and low-grade prostate cancer (HR = 1.22, 95% CI = 1.11 to 1.33). Men whose prostate cancer was treated with α1-adrenergic receptor antagonists underwent more frequent prostate-specific antigen testing. CONCLUSIONS Our findings show no significant association between α1-adrenergic receptor adrenoceptor antagonist exposure and prostate cancer mortality or high-grade prostate cancer. Although the preclinical evidence indicates a potential chemopreventive effect, this study's findings do not support it.
Collapse
Affiliation(s)
- Lars Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shirin Razdan
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Thorgerdur Palsdottir
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Markus Aly
- Department of Urology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tobias Nordström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Dara Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ash Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Lantz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Urology, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
2
|
Kang XC, Chen T, Zhou JL, Shen PY, Dai SH, Gao CQ, Zhang JY, Xiong XY, Liu DB. Phytosterols in hull-less pumpkin seed oil, rich in ∆ 7-phytosterols, ameliorate benign prostatic hyperplasia by lowing 5α-reductase and regulating balance between cell proliferation and apoptosis in rats. Food Nutr Res 2022; 65:7537. [PMID: 34984064 PMCID: PMC8693601 DOI: 10.29219/fnr.v65.7537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/28/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
Background Pumpkin seed oil is widely used to treat benign prostatic hyperplasia (BPH), a common disease in elder men. However, its active components and mechanism have remained to be elucidated. Objective The objective of the present study was to investigate the active components of pumpkin seed oil and its mechanism against BPH. Design Total phytosterol (TPS) was isolated from hull-less pumpkin (Cucurbita pepo L. var. Styriaca) seed oil and analyzed by gas chromatography/mass spectrometry (GC/MS). Three phytosterols were purified by preparative HPLC (high performance liquid chromatography) and confirmed by NMR (nuclear magnetic resonance). TPS (3.3 mg/kg body weight, 1 mL/day/rat) was administered intragastrically to the testosterone propionate-induced BPH rats for 4 weeks. The structure changes of prostate tissues were assessed by hematoxylin & eosin (H&E) staining. The expression of androgen receptor (AR) and steroid receptor coactivator 1 (SRC-1) was analyzed by immunohistochemistry, while that of 5α-reductase (5AR), apoptosis, or proliferation-related growth factors/proteins was detected by real-time quantitative polymerase chain reaction or western blotting. Results The ∆7-phytosterols in TPS reached up to 87.64%. Among them, 24β-ethylcholesta-7,22,25-trienol, 24β-ethylcholesta-7,25(27)-dien-3-ol, and ∆7-avenasterol were confirmed by NMR. TPS treatment significantly ameliorated the pathological prostate enlargement and restored histopathological alterations of prostate in BPH rats. It effectively suppressed the expressions of 5AR, AR, and coactivator SRC-1. TPS inhibited the expression of proliferation-related growth factor epidermal growth factor, whereas it increased the expressions of apoptosis-related growth factor/gene transforming growth factor-β1. The proliferation-inhibiting effect was achieved by decreasing the ERK (extracellular signal-regulated kinase) phosphorylation, while apoptosis was induced by Caspase 3 activation through JNK (c-Jun N-terminal kinase) and p38 phosphorylation. Conclusion TPS from hull-less pumpkin seed oil, with ∆7-phytosterols as its main ingredients, is a potential nutraceutical for BPH prevention.
Collapse
Affiliation(s)
- Xin-Cong Kang
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China.,Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China.,State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, P. R. China.,Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China.,These authors contributed equally to this work
| | - Tian Chen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China.,These authors contributed equally to this work
| | - Jia-Li Zhou
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China
| | - Peng-Yuan Shen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China
| | - Si-Hui Dai
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China
| | - Chang-Qing Gao
- Department of Laboratory Animals, Xiang Ya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Jia-Yin Zhang
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China.,Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China.,State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, P. R. China
| | - Xing-Yao Xiong
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China.,Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China.,Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Dong-Bo Liu
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, P. R. China.,Hunan Provincial Key Laboratory of Crop Germplasm Innovation and Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China.,State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, P. R. China.,Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China
| |
Collapse
|
3
|
Isaacs JT, Brennen WN, Christensen SB, Denmeade SR. Mipsagargin: The Beginning-Not the End-of Thapsigargin Prodrug-Based Cancer Therapeutics. Molecules 2021; 26:7469. [PMID: 34946547 PMCID: PMC8707208 DOI: 10.3390/molecules26247469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
Søren Brøgger Christensen isolated and characterized the cell-penetrant sesquiterpene lactone Thapsigargin (TG) from the fruit Thapsia garganica. In the late 1980s/early 1990s, TG was supplied to multiple independent and collaborative groups. Using this TG, studies documented with a large variety of mammalian cell types that TG rapidly (i.e., within seconds to a minute) penetrates cells, resulting in an essentially irreversible binding and inhibiting (IC50~10 nM) of SERCA 2b calcium uptake pumps. If exposure to 50-100 nM TG is sustained for >24-48 h, prostate cancer cells undergo apoptotic death. TG-induced death requires changes in the cytoplasmic Ca2+, initiating a calmodulin/calcineurin/calpain-dependent signaling cascade that involves BAD-dependent opening of the mitochondrial permeability transition pore (MPTP); this releases cytochrome C into the cytoplasm, activating caspases and nucleases. Chemically unmodified TG has no therapeutic index and is poorly water soluble. A TG analog, in which the 8-acyl groups is replaced with the 12-aminododecanoyl group, afforded 12-ADT, retaining an EC50 for killing of <100 nM. Conjugation of 12-ADT to a series of 5-8 amino acid peptides was engineered so that they are efficiently hydrolyzed by only one of a series of proteases [e.g., KLK3 (also known as Prostate Specific Antigen); KLK2 (also known as hK2); Fibroblast Activation Protein Protease (FAP); or Folh1 (also known as Prostate Specific Membrane Antigen)]. The obtained conjugates have increased water solubility for systemic delivery in the blood and prevent cell penetrance and, thus, killing until the TG-prodrug is hydrolyzed by the targeting protease in the vicinity of the cancer cells. We summarize the preclinical validation of each of these TG-prodrugs with special attention to the PSMA TG-prodrug, Mipsagargin, which is in phase II clinical testing.
Collapse
Affiliation(s)
- John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - William Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Samuel R. Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
4
|
Shah A, Shah AA, K N, Lobo R. Mechanistic targets for BPH and prostate cancer-a review. REVIEWS ON ENVIRONMENTAL HEALTH 2021; 36:261-270. [PMID: 32960781 DOI: 10.1515/reveh-2020-0051] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/02/2020] [Indexed: 05/26/2023]
Abstract
All men, almost, suffer from prostatic disorders in average life expectancy. In the year of 1950s, the first autopsy of prostate gland discovered the link between Benign prostatic hyperplasia (BPH) and Prostate Cancer (PCa). After that, many histology, biochemistry, epidemiology studies explained the association and associated risk factor for the same. From the various scientific evidence, it is proved that both diseases share some common transcription factors and signalling pathways. Still, BPH cannot be considered as the first step of PCa progression. To define, the relationship between both of the diseases, a well-defined large epidemiological study is needed. Along with androgen signalling, imbalanced apoptosis, oxidative stress, and microbial infection also crucial factors that significantly affect the pathogenesis of BPH. Various signalling pathways are involved in the progression of BPH. Androgen signalling is the driving force for the progress of PCa. In PCa androgen signalling is upregulated as compared to a healthy prostate. Some dominant Androgen-regulated genes and their functions have been discussed in this work.
Collapse
Affiliation(s)
- Abhishek Shah
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Aarti Abhishek Shah
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nandakumar K
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Richard Lobo
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
5
|
Cheon SY, Jin BR, Kim HJ, An HJ. Oleanolic Acid Ameliorates Benign Prostatic Hyperplasia by Regulating PCNA-Dependent Cell Cycle Progression In Vivo and In Vitro. JOURNAL OF NATURAL PRODUCTS 2020; 83:1183-1189. [PMID: 32191471 DOI: 10.1021/acs.jnatprod.9b01210] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oleanolic acid (OA) is a natural, biologically active pentacyclic triterpenoid found in Cornus officinalis. Although C. officinalis and OA have antiproliferative actions, the effects and mechanisms of OA in benign prostatic hyperplasia (BPH) are unclear. We examined the effect of OA in an animal model of testosterone-induced BPH. Male rats were injected with testosterone propionate with or without OA. The inhibitory effect of OA on BPH-1 cells was determined in vitro. Rats with BPH exhibited outstanding BPH symptoms, including prostatic enlargement, upregulated dihydrotestosterone and 5α-reductase 2 levels, and histological changes. Compared with the BPH group, the OA group showed fewer pathological alterations and regular androgen events. OA inhibited prostate cell proliferation by downregulating the expression of proliferating cell nuclear antigen (PCNA) and cell cycle markers in BPH-induced animals. This indicated that OA has superior therapeutic effect in the BPH animal model than finasteride. In vitro studies demonstrated upregulation of PCNA and cell cycle proteins, whereas OA clearly reduced this upregulation. Thus, OA may inhibit the development of BPH by targeting cell cycle progression markers. These suggest that OA is a potential agent for BPH treatment.
Collapse
Affiliation(s)
- Se-Yun Cheon
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Bo-Ram Jin
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| | - Hyo-Jung Kim
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sang-ji University, Wonju-si, Gangwon-do 26339, Republic of Korea
| |
Collapse
|
6
|
Chen P, Xiao H, Huang W, Xu DQ, Guo YM, Wang X, Wang XH, DiSanto ME, Zhang XH. Testosterone regulates myosin II isoforms expression and functional activity in the rat prostate. Prostate 2018; 78:1283-1298. [PMID: 30073674 DOI: 10.1002/pros.23702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/11/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is mainly caused by increased prostatic smooth muscle (SM) tone and prostatic volume. At the molecular level, SM myosin II (SMM II) and non-muscle myosin II (NMM II) mediate SM tone and cell proliferation while testosterone (T) plays a permissive role in the development of BPH. AIMS The novel objective of this study was to elucidate the effects of T on the proliferation and apoptosis of rat prostatic cells and SM contractility as well as related regulatory signaling pathways. MATERIALS AND METHODS Briefly, 36 male rats were divided into three groups (sham-operated, surgically castrated, and castrated with T supplementation). In vitro organ bath studies, competitive RT-PCR, Western-blotting analysis, Masson's trichrome staining, and immunofluorescence staining were performed. RESULTS Our data showed that castration dramatically increased prostatic SM contractility and SM MHC immunostaining revealed a relatively increased SM cell numbers in the stroma. T deprivation altered prostate SMM II isoform composition with upregulation of SM-B and SM2 but downregulation of LC17a, favoring a faster more phasic-type contraction. Moreover, protein expressions of MLCK, p-MLCP, RhoB, ROCK1, and ROCK2 increased in castrated rats. Meanwhile NMM II heavy chain isoforms A, B, and C (NMMHC-A, B, and C isoforms) were altered by castration which may be linked to decreased cell proliferation and increased apoptosis. CONCLUSION Our novel data demonstrated T regulates SMM II and NMM II and their functional activities in rat prostate and T ablation not only decreases prostate size (static component) but also changes the prostatic SM tone (dynamic component).
Collapse
Affiliation(s)
- Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Wei Huang
- Department of Urology, People's Hospital of Tuanfeng County, Hubei, China
| | - De-Qiang Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Yu-Ming Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Xiao Wang
- Department of Urology, People's Hospital of Wuhan University, Wuhan, China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Michael E DiSanto
- Departments of Biomedical Sciences and Surgery, Cooper Medical School of Rowan University, Camden, New Jersey
| | - Xin-Hua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
7
|
Kimbrough-Allah MN, Millena AC, Khan SA. Differential role of PTEN in transforming growth factor β (TGF-β) effects on proliferation and migration in prostate cancer cells. Prostate 2018; 78:377-389. [PMID: 29341212 PMCID: PMC5820153 DOI: 10.1002/pros.23482] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) acts as a tumor suppressor in normal epithelial cells but as a tumor promoter in advanced prostate cancer cells. PI3-kinase pathway mediates TGF-β effects on prostate cancer cell migration and invasion. PTEN inhibits PI3-kinase pathway and is frequently mutated in prostate cancers. We investigated possible role(s) of PTEN in TGF-β effects on proliferation and migration in prostate cancer cells. METHODS Expression of PTEN mRNA and proteins were determined using RT-PCR and Western blotting in RWPE1 and DU145 cells. We also studied the role of PTEN in TGF-β effects on cell proliferation and migration in DU145 cells after transient silencing of endogenous PTEN. Conversely, we determined the role of PTEN in cell proliferation and migration after over-expression of PTEN in PC3 cells which lack endogenous PTEN. RESULTS TGF-β1 and TGF-β3 had no effect on PTEN mRNA levels but both isoforms increased PTEN protein levels in DU145 and RWPE1 cells indicating that PTEN may mediate TGF-β effects on cell proliferation. Knockdown of PTEN in DU145 cells resulted in significant increase in cell proliferation which was not affected by TGF-β isoforms. PTEN overexpression in PC3 cells inhibited cell proliferation. Knockdown of endogenous PTEN enhanced cell migration in DU145 cells, whereas PTEN overexpression reduced migration in PC3 cells and reduced phosphorylation of AKT in response to TGF-β. CONCLUSION We conclude that PTEN plays a role in inhibitory effects of TGF-β on cell proliferation whereas its absence may enhance TGF-β effects on activation of PI3-kinase pathway and cell migration.
Collapse
Affiliation(s)
| | - Ana C Millena
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia
| | - Shafiq A Khan
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia
| |
Collapse
|
8
|
Ünlü A, Leake R. Transforming Growth Factor β1 Stimulates Urokinase Plasminogen Activator System on Prostate Cancer Cells. Int J Biol Markers 2018. [DOI: 10.1177/172460080301800208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effect of TGFβ1 on the proliferation and plasminogen activator system (PA) of two prostate carcinoma cell lines, PC3 and DU145, was investigated. PA, particularly urokinase plasminogen activator (uPA), has been implicated in extracellular proteolysis, local invasiveness, metastatic spread and angiogenesis. High levels of uPA and plasminogen activator inhibitor-1 (PAI-1) correlate with poor prognosis in several cancers. TGFβ1 had no significant effect on the proliferation of either cell line. TGFβ1 increased the production of uPA in PC3 and DU145 cells. Despite the very low PAI-1 protein levels in both cell lines, TGFβ1 treatment resulted in a remarkable increase in PAI-1 secretion. PAI-2 protein was also increased by 59% in the PC3 cells. A divergent effect of TGFβ1 on the uPA enzyme activity was observed (28% decrease in PC3 and 131% increase in DU145 cells). Overall, TGFβ1 treatment did not affect the invasion of reconstituted basement membrane of PC3 cells. In addition to the uPA:PAI-1 ratio, the presence of PAI-2 may be an important factor in the determination of metastatic sites for prostate cancer cells. In conclusion, the potential contribution of TGFβ1 to tumor invasion may be considered as positive, based on both loss of growth inhibition and stimulation of components of the invasive system of prostate carcinoma.
Collapse
Affiliation(s)
- A. Ünlü
- Department of Biochemistry, Medical Faculty, Mersin University, Mersin - Turkey
| | - R.E. Leake
- Department of Biochemistry, IBLS, Glasgow University, Glasgow - United Kingdom
| |
Collapse
|
9
|
Cynanchum wilfordii Ameliorates Testosterone-Induced Benign Prostatic Hyperplasia by Regulating 5α-Reductase and Androgen Receptor Activities in a Rat Model. Nutrients 2017; 9:nu9101070. [PMID: 28953224 PMCID: PMC5691687 DOI: 10.3390/nu9101070] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/17/2017] [Accepted: 09/21/2017] [Indexed: 11/17/2022] Open
Abstract
Benign prostatic hyperplasia (BPH) is characterized by uncontrolled proliferation of the prostate gland. Cynanchum wilfordii has been reported to improve sexual behavior in male rats. In this study, we investigated the protective effect of an aqueous extract of C. wilfordii (CWW) against BPH development in a testosterone-induced BPH rat model. The rats were divided into the following six groups: sham/vehicle; BPH/vehicle; BPH/finasteride; and three CWW doses (50, 100, and 200 mg/kg). After a 4-week treatment with CWW, the rats were euthanized at scheduled times, and their prostates were weighed, followed by a histopathological examination. Prostate growth inhibition rates in rats administered CWW 50, 100, and 200 mg/kg were 54.5%, 51.8%, and 50.1%, respectively. The BPH/CWW group showed decreased serum testosterone and dihydrotestosterone (DHT) levels compared to the BPH/vehicle group. Furthermore, the BPH/CWW group showed reduced prostate testosterone and DHT levels compared to the BPH/vehicle group. Mechanistically, the reverse transcription-polymerase chain reaction revealed downregulated mRNA expression levels of the androgen receptor, 5α-reductase, and B-cell lymphoma-2 (Bcl-2) in the BPH/CWW200 group compared with those in the testosterone-induced groups. In conclusion, these findings show the effectiveness of CWW in slowing the progression of testosterone-induced BPH in rats.
Collapse
|
10
|
Hu J, Wang G, Sun T. Dissecting the roles of the androgen receptor in prostate cancer from molecular perspectives. Tumour Biol 2017; 39:1010428317692259. [PMID: 28475016 DOI: 10.1177/1010428317692259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Androgen receptor plays a pivotal role in prostate cancer progression, and androgen deprivation therapy to intercept androgen receptor signal pathway is an indispensable treatment for most advanced prostate cancer patients to delay cancer progression. However, the emerging of castration-resistant prostate cancer reminds us the alteration of androgen receptor, which includes androgen receptor mutation, the formation of androgen receptor variants, and androgen receptor distribution in cancer cells. In this review, we introduce the process of androgen receptor and also its variants' formation, translocation, and function alteration by protein modification or interaction with other pathways. We dissect the roles of androgen receptor in prostate cancer from molecular perspective to provide clues for battling prostate cancer, especially castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jieping Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Muniyan S, Chaturvedi NK, Dwyer JG, LaGrange CA, Chaney WG, Lin MF. Human prostatic acid phosphatase: structure, function and regulation. Int J Mol Sci 2013; 14:10438-64. [PMID: 23698773 PMCID: PMC3676848 DOI: 10.3390/ijms140510438] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/08/2013] [Accepted: 05/08/2013] [Indexed: 11/16/2022] Open
Abstract
Human prostatic acid phosphatase (PAcP) is a 100 kDa glycoprotein composed of two subunits. Recent advances demonstrate that cellular PAcP (cPAcP) functions as a protein tyrosine phosphatase by dephosphorylating ErbB-2/Neu/HER-2 at the phosphotyrosine residues in prostate cancer (PCa) cells, which results in reduced tumorigenicity. Further, the interaction of cPAcP and ErbB-2 regulates androgen sensitivity of PCa cells. Knockdown of cPAcP expression allows androgen-sensitive PCa cells to develop the castration-resistant phenotype, where cells proliferate under an androgen-reduced condition. Thus, cPAcP has a significant influence on PCa cell growth. Interestingly, promoter analysis suggests that PAcP expression can be regulated by NF-κB, via a novel binding sequence in an androgen-independent manner. Further understanding of PAcP function and regulation of expression will have a significant impact on understanding PCa progression and therapy.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mails: (S.M.); (N.K.C.); (W.G.C.)
| | - Nagendra K. Chaturvedi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mails: (S.M.); (N.K.C.); (W.G.C.)
| | - Jennifer G. Dwyer
- College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mail:
| | - Chad A. LaGrange
- Department of Surgery/Urology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mail:
| | - William G. Chaney
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mails: (S.M.); (N.K.C.); (W.G.C.)
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mails: (S.M.); (N.K.C.); (W.G.C.)
- Department of Surgery/Urology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; E-Mail:
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
12
|
Danielpour D. Transforming Growth Factor-Beta in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
13
|
Human Prostatic Acid Phosphatase in Prostate Carcinogenesis. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
14
|
Morse MD, McNeel DG. T cells localized to the androgen-deprived prostate are TH1 and TH17 biased. Prostate 2012; 72:1239-47. [PMID: 22213030 PMCID: PMC3673717 DOI: 10.1002/pros.22476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/28/2011] [Indexed: 12/20/2022]
Abstract
BACKGROUND T cells infiltrate the prostates of prostate cancer patients undergoing neoadjuvant androgen deprivation. These prostate-infiltrating T cells have an oligoclonal phenotype, suggesting the development of an antigen-specific T-cell response. We hypothesized that androgen deprivation might elicit a prostate tissue-specific T-cell response that could potentially be combined with other immune-active therapies, and consequently sought to investigate the nature and timing of this T-cell response following castration. METHODS We investigated the phenotype and cytokine expression of T cells at various time points in the prostates of Lewis rats following surgical castration, and used adoptive transfer of prostate-infiltrating lymphocytes (PILs) to determine whether the infiltration by T cells was mediated by effects of castration on the prostate or lymphocytes. RESULTS Prostate T-cell infiltration shortly after castration was T(H) 1 biased up to approximately 30 days, followed by a predominance of T(H) 17-type cells, which persisted until at least 90 days post castration. PILs from sham-treated or castrate rats localized to the prostates of castrate animals. CONCLUSIONS These observations suggest castration elicits a time-dependent prostate-specific T-cell infiltration, and this infiltration is likely mediated by effects of castration on prostate tissue rather than T-cells. These findings have implications for the timing of immunotherapies combined with androgen deprivation as treatments for prostate cancer.
Collapse
Affiliation(s)
- Matthew D. Morse
- Department of Cancer Biology, University of Wisconsin, Madison
- Department of Medicine, University of Wisconsin, Madison
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin, Madison
- To whom correspondence should be addressed: 7007 Wisconsin Institutes of Medical Research, 1111 Highland Avenue, Madison, WI 53705. Tel: (608) 265-8131 Fax: (608) 265-0614
| |
Collapse
|
15
|
Grosse A, Bartsch S, Baniahmad A. Androgen receptor-mediated gene repression. Mol Cell Endocrinol 2012; 352:46-56. [PMID: 21784131 DOI: 10.1016/j.mce.2011.06.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Revised: 06/21/2011] [Accepted: 06/27/2011] [Indexed: 11/19/2022]
Abstract
Androgens have an essential role in inducing the genetic program for masculinization during development. Androgens mediate their effect through the androgen receptor (AR), a ligand-controlled transcription factor and regulator of rapid signaling. Inactivated AR results in complete feminization. Androgens are also essential in later life for reproduction, behavior, muscle development, breast, and prostate growth. In general, androgens inhibit breast and promote prostate growth. In the latter context the AR is a major drug target. On the one hand, many insights have been obtained how the AR mediates gene activation on a molecular level. Gene activation is mediated by a battery of factors including coactivators, chromatin remodeling complex proteins and transcription factors which either directly or indirectly interact with the AR at DNA binding sites. On the other hand, there are important AR target genes that are repressed by androgen-bound AR. However, the underlying molecular mechanisms are poorly understood although genes repressed by AR are key factors involved in cell proliferation and invasion. Here, we summarize molecular mechanisms of AR-mediated gene repression, thereby differentiating between direct and indirect DNA/chromatin recruitment and between genomic and non-genomic effects.
Collapse
Affiliation(s)
- Andreas Grosse
- Institute of Human Genetics, Jena University Hospital, D-07743 Jena, Germany
| | | | | |
Collapse
|
16
|
Lai KP, Yamashita S, Vitkus S, Shyr CR, Yeh S, Chang C. Suppressed prostate epithelial development with impaired branching morphogenesis in mice lacking stromal fibromuscular androgen receptor. Mol Endocrinol 2011; 26:52-66. [PMID: 22135068 DOI: 10.1210/me.2011-1189] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Using the cre-loxP system, we generated a new mouse model [double stromal androgen receptor knockout (dARKO)] with selectively deleted androgen receptor (AR) in both stromal fibroblasts and smooth muscle cells, and found the size of the anterior prostate (AP) lobes was significantly reduced as compared with those from wild-type littermate controls. The reduction in prostate size of the dARKO mouse was accompanied by impaired branching morphogenesis and partial loss of the infolding glandular structure. Further dissection found decreased proliferation and increased apoptosis of the prostate epithelium in the dARKO mouse AP. These phenotype changes were further confirmed with newly established immortalized prostate stromal cells (PrSC) from wild-type and dARKO mice. Mechanistically, IGF-1, placental growth factor, and secreted phosphoprotein-1 controlled by stromal AR were differentially expressed in PrSC-wt and PrSC-ARKO. Moreover, the conditioned media (CM) from PrSC-wt promoted prostate epithelium growth significantly as compared with CM from PrSC-dARKO. Finally, adding IGF-1/placental growth factor recombinant proteins into PrSC-dARKO CM was able to partially rescue epithelium growth. Together, our data concluded that stromal fibromuscular AR could modulate epithelium growth and maintain cellular homeostasis through identified growth factors.
Collapse
Affiliation(s)
- Kuo-Pao Lai
- George H Whipple Laboratory for Cancer Research, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | |
Collapse
|
17
|
Jiang Q, Han BM, Zhao FJ, Hong Y, Xia SJ. The differential effects of prostate stromal cells derived from different zones on prostate cancer epithelial cells under the action of sex hormones. Asian J Androl 2011; 13:798-805. [PMID: 21765438 DOI: 10.1038/aja.2011.22] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is well known that prostate cancer (PCa) occurs predominantly in the peripheral zone (PZ), whereas benign prostatic hyperplasia (BPH) typically develops in the transition zone. To identify possible mechanisms underlying zonal differences, we compared the effects of prostate stromal cells derived from the peripheral zone (PZsc) and the transition zone (TZsc) on a PCa epithelial cell line (PC3) in the presence of sex hormones. First, we observed that androgen receptor (AR) mRNA was more highly expressed in PZsc than TZsc when the cells were treated with dihydrotestosterone (DHT) and β-oestradiol (E2) (P<0.05). By ELISA, we looked for differences in the secretion of peptide growth factors from PZsc and TZsc. We found that keratinocyte growth factor (KGF) secretion increased with increasing concentrations of DHT (P<0.01) and was higher in PZsc than TZsc. Under treatment with DHT plus E2, PZsc secreted more transforming growth factor-β1 (TGF-β1) than TZsc, but this pattern was reversed when the cells were treated with E2 only. With increasing concentrations of DHT, insulin-like growth factor-1 (IGF-1) secretion increased in PZsc but decreased in TZsc. To further characterize the effects of PZsc and TZsc on PC3 cells, we developed a coculture model and performed MTT assays, Western blot analysis and real-time RT-PCR. We found that PZsc promoted PC3 cell proliferation and progression better than TZsc, particularly when treated with 10 nmol l(-1) DHT plus 10 nmol l(-1) E2. In conclusion, our data suggest that PZsc may have a greater capacity to induce PCa development and progression than TZsc via growth factors regulated by sex hormones. These findings provide possible mechanisms underlying zonal differences in prostate diseases, which may aid the search for novel therapeutic targets for PCa.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Urology, Shanghai First People's Hospital, Institute of Urology, Shanghai Jiao Tong University, Shanghai 200080, China
| | | | | | | | | |
Collapse
|
18
|
Davis JS, Nastiuk KL, Krolewski JJ. TNF is necessary for castration-induced prostate regression, whereas TRAIL and FasL are dispensable. Mol Endocrinol 2011; 25:611-20. [PMID: 21292828 DOI: 10.1210/me.2010-0312] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
TNF, a proinflammatory and immune-regulatory cytokine, is a potent apoptotic stimulus in vitro. However, there have been few examples of a physiologic role for TNF-induced apoptosis in vivo. Here, we describe a novel role for TNF in prostate epithelial cell apoptosis after androgen withdrawal. Employing high-resolution serial magnetic resonance imaging to measure mouse prostate volume changes over time, we demonstrate that the extent of castration-induced prostate regression is significantly reduced in mice null for either the Tnf or Tnfr1 genes but not mice deficient for TNF-related apoptosis-inducing ligand or Fas signaling. Wild-type mice receiving soluble TNF (sTNF) receptor 2 (to bind TNF and block signaling) before castration exhibit an identical reduction of prostate regression. Together, these data indicate that uniquely among known extrinsic death signals, TNF is required for castration-induced prostate regression. Additionally, membrane-bound TNF protein and stromal cell specific TNF mRNA levels increase in rat prostate after castration. This is consistent with a paracrine role for TNF in prostate regression. When injected into the peritoneum of Tnf(-/-) mice at the time of castration, sTNF restores normal levels of prostate regression. However, wild-type mice receiving sTNF in the absence of castration do not exhibit prostate regression, indicating that TNF alone is not sufficient but acts in the context of additional castration-induced signals. These findings support a physiologic role for TNF in prostate regression after androgen withdrawal. Understanding this role may lead to novel therapies for prostate cancer.
Collapse
Affiliation(s)
- Jennifer S Davis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, California 92697-4800, USA
| | | | | |
Collapse
|
19
|
Moses H, Barcellos-Hoff MH. TGF-beta biology in mammary development and breast cancer. Cold Spring Harb Perspect Biol 2011; 3:a003277. [PMID: 20810549 DOI: 10.1101/cshperspect.a003277] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transforming growth factor-β1 (TGF-β) was first implicated in mammary epithelial development by Daniel and Silberstein in 1987 and in breast cancer cells and hormone resistance by Lippman and colleagues in 1988. TGF-β is critically important for mammary morphogenesis and secretory function through specific regulation of epithelial proliferation, apoptosis, and extracellular matrix. Differential TGF-β effects on distinct cell types are compounded by regulation at multiple levels and the influence of context on cellular responses. Studies using controlled expression and conditional-deletion mouse models underscore the complexity of TGF-β biology across the cycle of mammary development and differentiation. Early loss of TGF-β growth regulation in breast cancer evolves into fundamental deregulation that mediates cell interactions and phenotypes driving invasive disease. Two outstanding issues are to understand the mechanisms of biological control in situ and the circumstances by which TGF-β regulation is subverted in neoplastic progression.
Collapse
Affiliation(s)
- Harold Moses
- Department of Cancer Biology and Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
20
|
Reis STD, Pontes-Júnior J, Antunes AA, Sousa-Canavez JMD, Abe DK, Cruz JASD, Dall'oglio MF, Crippa A, Passerotti CC, Ribeiro-Filho LA, Viana NI, Srougi M, Leite KRM. Tgf-β1 expression as a biomarker of poor prognosis in prostate cancer. Clinics (Sao Paulo) 2011; 66:1143-7. [PMID: 21876965 PMCID: PMC3148455 DOI: 10.1590/s1807-59322011000700004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 03/13/2011] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To evaluate the correlation between transforming growth factor beta (TGF-β1) expression and prognosis in prostate cancer. PATIENTS AND METHODS TGF-β1 expression levels were analyzed using the quantitative real-time polymerase chain reaction to amplify RNA that had been isolated from fresh-frozen malignant and benign tissue specimens collected from 89 patients who had clinically localized prostate cancer and had been treated with radical prostatectomy. The control group consisted of li patients with benign prostate hyperplasia. The expression levels of TGF-β1 were compared between the groups in terms of Gleason scores, pathological staging, and prostate-specific antigen serum levels. RESULTS In the majority of the tumor samples, TGF-β1 was underexpressed 67.0% of PCa patients. The same expression pattern was identified in benign tissues of patients with prostate cancer. Although most cases exhibited underexpression of TGF-β1, a higher expression level was found in patients with Gleason scores ≥ 7 when compared to patients with Gleason scores < 7(p = 0.002). Among the 26 cases of TGF-β1 overexpression, 92.3% had poor prognostic features. CONCLUSIONS TGF-β1 was underexpressed in prostate cancers; however, higher expression was observed in tumors with higher Gleason scores, which suggests that TGF-β1 expression may be a useful prognostic marker for prostate cancer. Further studies of clinical specimens are needed to clarify the role of TGF-β1 in prostate carcinogenesis.
Collapse
Affiliation(s)
- Sabrina Thalita Dos Reis
- Laboratory of Medical Investigation (LIM55), Urology Department, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Reis STD, Pontes-Júnior J, Antunes AA, Sousa-Canavez JMD, Abe DK, Cruz JASD, Dall'oglio MF, Crippa A, Passerotti CC, Ribeiro-Filho LA, Viana NI, Srougi M, Leite KRM. Tgf-β1 expression as a biomarker of poor prognosis in prostate cancer. Clinics (Sao Paulo) 2011. [PMID: 21876965 DOI: 10.1590/s1807-59322011000700004.pmid:21876965;pmcid:pmc3148455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
OBJECTIVE To evaluate the correlation between transforming growth factor beta (TGF-β1) expression and prognosis in prostate cancer. PATIENTS AND METHODS TGF-β1 expression levels were analyzed using the quantitative real-time polymerase chain reaction to amplify RNA that had been isolated from fresh-frozen malignant and benign tissue specimens collected from 89 patients who had clinically localized prostate cancer and had been treated with radical prostatectomy. The control group consisted of li patients with benign prostate hyperplasia. The expression levels of TGF-β1 were compared between the groups in terms of Gleason scores, pathological staging, and prostate-specific antigen serum levels. RESULTS In the majority of the tumor samples, TGF-β1 was underexpressed 67.0% of PCa patients. The same expression pattern was identified in benign tissues of patients with prostate cancer. Although most cases exhibited underexpression of TGF-β1, a higher expression level was found in patients with Gleason scores ≥ 7 when compared to patients with Gleason scores < 7(p = 0.002). Among the 26 cases of TGF-β1 overexpression, 92.3% had poor prognostic features. CONCLUSIONS TGF-β1 was underexpressed in prostate cancers; however, higher expression was observed in tumors with higher Gleason scores, which suggests that TGF-β1 expression may be a useful prognostic marker for prostate cancer. Further studies of clinical specimens are needed to clarify the role of TGF-β1 in prostate carcinogenesis.
Collapse
Affiliation(s)
- Sabrina Thalita Dos Reis
- Laboratory of Medical Investigation (LIM55), Urology Department, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhao Y, Peng J, Zheng L, Yu W, Jin J. Transforming growth factor beta1 mediates apoptotic activity of angiotensin II type I receptor blocker on prostate epithelium in vitro. Prostate 2010; 70:899-905. [PMID: 20135646 DOI: 10.1002/pros.21124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The significant association of benign prostatic hyperplasia (BPH) and hypertension indicates a common pathophysiological factor for both diseases. Hyperactivity of the renin-angiotensin system (RAS) has been reported in BPH. Angiotensin II type I (AT1) receptor is the principal mediator of the RAS, and the antagonist, AT1 receptor blocker (ARB), can induce apoptosis in prostate epithelium cells and increase transforming growth factor beta1 (TGF-beta1) expression. We aimed to investigate the mechanism of inhibition of AT1 receptor in prostate epithelium cells and the role of TGF-beta1. METHODS Human prostate epithelium cell lines were treated with different concentrations of ARB (losartan) (0, 0.1, 1, 10, 100, and 1,000 microM) for 24-72 hr. Cell proliferation was analyzed by cell proliferation assay. The location of AT1 receptor was shown by immunocytohistochemistry and immunocytofluorescence study. Analysis of apoptosis was by use of terminal transferase TdT-mediated dUTP-biotin end labeling (TUNEL) and caspase 3/7 activity assay. Mitochondrial outer-membrane permeabilization was measured by JC-1 staining. The level of TGF-beta1 was determined by enzyme-linked immunosorbent assay. RESULTS Immunohistochemistry and immunofluorescence analysis showed AT1 receptor expressed in epithelium cells. Compared to control cultures, cultures treated with losartan for 24-72 hr showed a dose-dependent significant decrease in cell number, with apoptosis increased by 65.2%. Decreased cell number was reversed on treatment with anti-TGF-beta1 antibody. TUNEL staining showed increased apoptosis in prostate epithelium cells exposed to losartan. Caspase 3/7 activation was increased and mitochondrial membrane potential was downregulated. Expression of TGF-beta1 in cells treated with losartan was higher than that in untreated cells. CONCLUSIONS The apoptotic effect of blockade of AT1 receptor on human prostatic epithelium cells may be mediated through an autocrine the production of TGF-beta1. Furthermore, this finding may have implications for medication options. Inc.
Collapse
Affiliation(s)
- Yayuan Zhao
- Department of Urology, Peking University First Hospital, Beijing, China.
| | | | | | | | | |
Collapse
|
23
|
Diener KR, Need EF, Buchanan G, Hayball JD. TGF-beta signalling and immunity in prostate tumourigenesis. Expert Opin Ther Targets 2010; 14:179-92. [PMID: 20055717 DOI: 10.1517/14728220903544507] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
IMPORTANCE OF THE FIELD The TGF-beta's are pleiotropic cytokines that regulate multiple cellular functions. Their role in the prostate is important for normal prostate development and also in prostate tumourigenesis. AREAS COVERED IN THIS REVIEW The interactions TGF-beta-mediated signalling has with maintaining prostate health, as well as its role in prostate tumourigenesis and prostate tumour immune evasion, with emphasis on how a breakdown in these interactions may influence disease progression. WHAT THE READER WILL GAIN That TGF-beta influences normal prostate growth and differentiation by regulating the balance between epithelial cell proliferation and apoptosis, and involving the androgen receptor pathway. That TGF-beta protects and maintains prostate stem cells and a review of the contrasting role TGF-beta has in prostate tumourigenesis and tumour development, where TGF-beta acts as a tumour suppressor and then switches roles to become a tumour promoter, and creates a local immunosuppressive niche leading to systemic tumour tolerance. TAKE HOME MESSAGE TGF-beta signalling in prostate cancer is a valid target for the treatment of this disease; however any therapeutic regimen will require an understanding of all aspects of the TGF-beta-signalling nexus, otherwise by the very pleiotrophic nature of TGF-beta, limited clinical benefits may result.
Collapse
Affiliation(s)
- Kerrilyn R Diener
- Hanson Institute, Experimental Therapeutics Laboratory, Adelaide, SA 5000, Australia
| | | | | | | |
Collapse
|
24
|
Action, localization and structure-function relationship of growth factors and their receptors in the prostate. ACTA ACUST UNITED AC 2009. [DOI: 10.1017/s0962279900001265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Whereas the direct action of sex steroids, namely of androgens, on prostate cell division was questioned as early as in the 1970s, and remains so, the interest in prostatic growth factors (GFs) is rather recent but has expanded tremendously in the last five years. This lag period can be partly explained by the fact that, at the time, androgen receptors had just been discovered, and newly developed hormonal regimens or strategies to treat patients with prostate carcinoma (PCa) or epithelioma had generated great enthusiasm and hopes in the medical and scientific community. Another point to consider was the difficulty in maintaining prostate tissues in organ cultures and the relative novelty of culturing prostate epithelial cells in monolayers. Failures of sex steroids to elicit a direct positive response on prostate cell divisionin vitro, as seenin vivo, were interpreted as resulting from inappropriate models or culture conditions. However, the increasing number of reports confirming the lack of mitogenic activity of sex steroidsin vitro, coupled with the powerful mitogenic activity of GFs displayed in other systems, the discovery of GF receptors (GF-Rs), and the elucidation of their signalling pathways showing sex steroid receptors as potential substrates of GF-activated protein kinases gradually led to an increased interest in the putative role of GFs in prostate physiopathology. Of utmost importance was the recognition that hormone refractiveness was responsible for PCa progression, and for the poor outcome of patients with advanced disease under endocrine therapies. This problem remains a major issue and it raises several key questions that need to be solved at the fundamental and clinical levels.
Collapse
|
25
|
Jones E, Pu H, Kyprianou N. Targeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progression. Expert Opin Ther Targets 2009; 13:227-34. [PMID: 19236240 DOI: 10.1517/14728220802705696] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND TGF-beta regulates prostate growth by inhibiting epithelial cell proliferation and inducing apoptosis through eliciting a dynamic signaling pathway. In metastatic prostate cancer, however, TGF-beta serves as a tumor promoter. TGF-beta engages Smad-dependent and Smad-independent mechanisms to exert its action. During prostate tumorigenesis, prostate cells exhibit loss or mutation of TGF-beta transmembrane receptors. Increased production of TGF-beta causes immunosuppression, extracellular matrix degradation, epithelia to mesenchymal transition and angiogenesis that promotes tumor cell invasion and metastasis. OBJECTIVE The molecular basis for effective therapeutic targeting of TGF-beta must be directed towards the double-edge-sword nature of the cytokine: inhibiting the TGF-beta tumor promoter capabilities in advanced metastatic prostate cancer, although retaining the growth-inhibitory abilities exhibited in early stages of prostate tumorigenesis. RESULTS/CONCLUSION The current understanding of the therapeutic possibilities of targeting TGF-beta signaling during prostate tumor progression is built on preclinical studies. Studies targeting TGF-beta signaling pathway for the treatment of several human malignancies include the use of neutralizing antibodies, antisense oligonucelotides and small molecule inhibitors of kinase activity of the receptor complex. This review focuses on exploiting the therapeutic potential of targeting TGF-beta signaling in the context of its contribution to prostate cancer initiation and progression to metastasis.
Collapse
Affiliation(s)
- Elisabeth Jones
- University of Kentucky, Surgery/Urology and Markey Cancer Center, 306 Combs Building, 800 Rose Street, Lexington, KY 40536, USA
| | | | | |
Collapse
|
26
|
Abstract
The epidemiology of benign prostatic hyperplasia (BPH) is complex and not fully understood. The androgenic hormones testosterones and dihydrotestosterone play at least a permissive and important role. Growth factors and other hormones including estrogens may also play a role. BPH is a truely hyperplastic process resulting in growth of glandular-epithelial and stromal/muscle tissue in the prostate, leading to often measurable growth taking on different shapes and configurations which may impact symptoms and secondary outcomes. It is important to recognize that BPH is a histological conditions, which is one but not the only cause of lower urinary tract symptoms, and may or may not be associated with prostate enlargement and bladder outlet obstruction. Recognizing the different entities and determining their presence in individual patients may help with therapeutic decision making.
Collapse
|
27
|
Yoo KS, Nastiuk KL, Krolewski JJ. Transforming growth factor ß1 induces apoptosis by suppressing FLICE-like inhibitory protein in DU145 prostate epithelial cells. Int J Cancer 2009; 124:834-42. [DOI: 10.1002/ijc.24024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
28
|
Abstract
TGF (transforming growth factor)-beta1 is a multifunctional cytokine that influences homoeostatic processes of various tissues. TGF-beta1 expression is inhibited by androgens in the prostate gland, whereas its expression is enhanced by androgens in highly metastatic prostate cancer cells. Here, we examined regulation of human TGF-beta1 promoter activity by androgen in prostate cancer cells. The full-length (-3363 to +110) promoter showed a high level of activity in response to androgen in PC3mm2 cells expressing AR (androgen receptor). Further deletion analysis revealed three distal and three proximal AREs (androgen-response elements) in the promoter. Gel-shift and footprint assays show that these AREs physically interact with the DNA-binding domain of AR. Chromatin immunoprecipitation assays revealed the androgen-dependent recruitment of AR to the ARE-containing regions of the TGF-beta1 gene. More importantly, a negative ARE was detected in the TGF-beta1 promoter. Both positive and negative AREs are functional in the androgen-regulated transcription of the TGF-beta1 promoter. These findings imply that androgen signalling may positively or negatively regulate TGF-beta1 expression in response to various signals or under different environmental conditions.
Collapse
|
29
|
Abstract
Stromal-epithelial interactions mediated by paracrine signaling mechanisms dictate prostate development and progression of prostate cancer. The regulatory role of androgens in both the prostate stromal and epithelial compartments set the prostate apart from many other organs and tissues with regard to gene targeting. The identification of androgen-dependent prostate epithelial promoters has allowed successful gene targeting to the prostate epithelial compartment. Currently, there are no transgenic mouse models available to specifically alter gene expression within the prostate stromal compartment. As a primary metastatic site for prostate cancer is bone, the functional dissection of the bone stromal compartment is important for understanding stromal-epithelial interactions associated with metastatic tumor growth. Use of currently available methodologies for the expression or deletion of gene expression in recent research studies has advanced our understanding of the stroma. However, the complexity of stromal heterogeneity within the prostate remains a challenge to obtaining compartment or cell-lineage-specific in vivo models necessary for furthering our understanding of prostatic developmental, benign, tumorigenic, and metastatic growth.
Collapse
Affiliation(s)
- Roger S Jackson
- Department of Urologic Surgery, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232-2765, USA
| | | | | |
Collapse
|
30
|
Abstract
The clinical dilemma today in the management of prostate cancer (PCA) is to distinguish men who need definitive treatment from men who have indolent disease. As demonstrated most recently by the randomized Scandinavian trial evaluating the benefit of prostatectomy over Watchful Waiting, surgery significantly decreased the risk of death from PCA. However, this same study also suggests that 19 men need to be treated to benefit one man. Given the high prevalence of the disease, the aging of the population, and the potential morbidity of treatment, the ability to distinguish aggressive from indolent forms of PCA is critical. Treatment for advanced PCA begins with androgen ablation, but eventually hormone-refractory (HR) PCA emerges. Novel therapies are in various stages of clinical trials, including kinase inhibitors, antisense oligonucleotides, and inhibitors of heat-shock proteins. The discovery of novel therapeutic approaches is an active area of clinical research. Eliminating HR PCA before it advances is a high priority in the biomarker field. Therefore, the development of molecular signatures of lethal PCA are critical. In addition, the recent discovery that a significant percentage of PCAs harbor a TMPRSS2-ETS gene fusion suggests that targeting either the ETS transcription factors or the fusion product may offer a novel approach to therapy. However, in 2007, the mainstay of treatment for advanced PCA remains androgen ablation therapy as originally introduced in the early 1940s.
Collapse
|
31
|
Yamashita S, Takahashi S, McDonell N, Watanabe N, Niwa T, Hosoya K, Tsujino Y, Shirai T, Ushijima T. Methylation silencing of transforming growth factor-beta receptor type II in rat prostate cancers. Cancer Res 2008; 68:2112-21. [PMID: 18381416 DOI: 10.1158/0008-5472.can-07-5282] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To identify methylation-silenced genes in prostate cancers, a microarray analysis for genes up-regulated by treatment with a demethylating agent, 5-aza-2'-deoxycytidine, was performed using three rat prostate cancer cell lines. Eight genes (Aebp1, Dysf, Gas6, LOC361288, Nnat, Ocm, RGD1308119, and Tgfbr2) were re-expressed at 16-fold or more, and their promoter CpG islands were shown to be densely methylated in the cancer cell lines. From the eight genes, Tgfbr2, a key mediator of transforming growth factor-beta (TGF-beta) signaling that has been strongly implicated in human and rat prostate carcinogenesis, was selected, and its silencing in primary samples was analyzed further. Tgfbr2 was methylated and markedly down-regulated in three of seven 3,2'-dimethyl-4-aminobiphenyl-induced invasive adenocarcinomas in the dorsolateral lobe of the rat prostate. In humans, marked down-regulation of TGFBR2 protein was observed in 12 of 20 high-grade prostatic intraepithelial neoplasia and 36 of 60 prostate cancers. DNA methylation of the human TGFBR2 promoter CpG islands repressed transcription, if present, but neither methylation nor mutation were detected in 27 human prostate cancers analyzed. Methylation silencing of rat Tgfbr2 was associated with histone H3 lysine 9 trimethylation, whereas decreased expression of human TGFBR2 was mainly due to decreased transcription activity, sometimes in concert with histone deacetylation and H3 lysine 27 trimethylation. The identification of methylation silencing of Tgfbr2 in rat prostate cancers, in accordance with TGFBR2 down-regulation in human prostate cancers, will enable us to analyze how aberrant methylation is induced in vivo and identify factors that promote and suppress the induction of aberrant methylation.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Carcinogenesis Division, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
FOXO3a mediates the androgen-dependent regulation of FLIP and contributes to TRAIL-induced apoptosis of LNCaP cells. Oncogene 2008; 27:4422-33. [DOI: 10.1038/onc.2008.80] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
Nastiuk KL, Yoo K, Lo K, Su K, Yeung P, Kutaka J, Danielpour D, Krolewski JJ. FLICE-Like Inhibitory Protein Blocks Transforming Growth Factor β1–Induced Caspase Activation and Apoptosis in Prostate Epithelial Cells. Mol Cancer Res 2008; 6:231-42. [DOI: 10.1158/1541-7786.mcr-07-0386] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Doxazosin reduces cell proliferation and increases collagen fibers in rat prostatic lobes. Cell Tissue Res 2008; 332:171-83. [DOI: 10.1007/s00441-007-0559-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Accepted: 11/15/2007] [Indexed: 10/22/2022]
|
35
|
|
36
|
Lambert JR, Kelly JA, Shim M, Huffer WE, Nordeen SK, Baek SJ, Eling TE, Lucia MS. Prostate derived factor in human prostate cancer cells: gene induction by vitamin D via a p53-dependent mechanism and inhibition of prostate cancer cell growth. J Cell Physiol 2006; 208:566-74. [PMID: 16741990 DOI: 10.1002/jcp.20692] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The secosteroid hormone 1alpha, 25-dihydroxyvitamin D3 (1,25D) has been shown to regulate the growth and differentiation of human prostate cancer (PCa) cells, although the precise molecular mechanisms mediating these effects have not been defined. Previous studies in our laboratory demonstrated that the antiproliferative effects of 1,25D on PCa cells are mediated through the nuclear vitamin D receptor (VDR). In the present study, we performed gene profiling of LNCaP human PCa cells following 1,25D treatment and identified the antitumorigenic gene, prostate derived factor (PDF), as being highly induced by 1,25D. PDF is a member of the TGF-beta superfamily and has been implicated in a variety of functions directly related totumorigenicity including antiproliferative and pro-apoptotic effects. Gene expression studies using 1,25D analogs and a VDR antagonist demonstrate that 1,25D-mediated induction of PDF message and protein in PCa cells is dependent on VDR action. PDF is a transcriptional target of the tumor suppressor, p53. Here we show that the expression of PDF in nine PCa cell lines is dependent on functional p53. Additionally, transfection of p53-null ALVA-31 PCa cells with a p53 expression plasmid, and expression of dominant negative p53 in LNCaP PCa cells, show that the ability of VDR to induce PDF requires functional p53. Importantly, forced PDF expression in PC-3 cells results in decreased cell proliferation, soft agar cloning, and xenograft tumor size. These data demonstrate that PDF exerts antitumorigenic properties on PCa cells and its regulation by 1,25D may provide insights into the action of 1,25D in PCa.
Collapse
Affiliation(s)
- James R Lambert
- Department of Pathology, University of Colorado Health Sciences Center, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Fukamachi H. Runx3 controls growth and differentiation of gastric epithelial cells in mammals. Dev Growth Differ 2006; 48:1-13. [PMID: 16466388 DOI: 10.1111/j.1440-169x.2006.00832.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Runx3 is a transcription factor expressed by gastric epithelial cells. In the Runx3(-/-) mouse, gastric epithelia exhibited hyperplasia, and epithelial apoptosis was suppressed. By analyzing growth of the epithelial cells in primary culture, we found that Runx3(-/-) gastric epithelial cells are less sensitive to the growth-inhibitory and apoptosis-inducing activities of TGF-beta, suggesting that Runx3 is a major growth regulator of gastric epithelial cells by regulating their response to TGF-beta. We also found that Runx3 plays an important role in the control of gastric epithelial differentiation. When subcutaneously implanted into nude mice, Runx3(-/-) gastric epithelial cells formed tumors in which some cells differentiated into intestinal-type cells. Clonal analysis showed that gastric epithelial cells transdifferentiate into intestinal-type cells in the tumor. Considering that gastric epithelial differentiation is very stable, and that intestinal-type cells never differentiate in the mouse stomach, it is remarkable that gastric epithelial cells transdifferentiate into intestinal-type cells. We conclude that Runx3 is deeply involved in the control of both growth and differentiation of gastric epithelial cells. The role of Runx3 in the specification of gastric epithelial cells is discussed.
Collapse
Affiliation(s)
- Hiroshi Fukamachi
- Department of Molecular Oncology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8519, Japan.
| |
Collapse
|
39
|
Mirone V, Fusco F, Verze P, Schulman C, Debruyne F, Imbimbo C. Androgens and Benign Prostatic Hyperplasia. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.eursup.2006.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
40
|
Perttu MC, Martikainen PM, Huhtala HSA, Bläuer M, Tammela TLJ, Tuohimaa PJ, Syvälä H. Altered levels of Smad2 and Smad4 are associated with human prostate carcinogenesis. Prostate Cancer Prostatic Dis 2006; 9:185-9. [PMID: 16550207 DOI: 10.1038/sj.pcan.4500871] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alterations have been demonstrated in ligand and cognate receptor system of the transforming growth factor beta (TGF-beta) pathway in prostate cancer (PC). Still, little is known about changes in the activity of the intracellular Smad cascade of TGF-beta signaling during prostate carcinogenesis. We used immunohistochemistry to analyze phosphorylated Smad2 (p-Smad2), nuclear Smad4 and inhibitory-Smad7 in epithelial cells of normal, hyperplastic and malignant prostate. Specimens comprised 49 tissue cores of PC, 10 benign prostate hypertrophies and three normal prostates. Nuclear p-Smad2 (P<0.001) and nuclear Smad4 (P=0.023) were significantly decreased in PC with remarkable variations in cytoplasmic Smad7 levels. Substantial decreases in p-Smad2 and Smad4 levels were found in specimens with primary Gleason grades 3 and 4, whereas in grade 5, levels were markedly higher. Our results provide the first evidence for changes and reversible attenuation in the Smad system of the TGF-beta pathway during prostate carcinogenesis.
Collapse
Affiliation(s)
- M C Perttu
- Department of Anatomy, Medical School, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | |
Collapse
|
41
|
Setlur SR, Rubin MA. Current thoughts on the role of the androgen receptor and prostate cancer progression. Adv Anat Pathol 2005; 12:265-70. [PMID: 16210922 DOI: 10.1097/01.pap.0000184179.50672.f7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Androgens play a central role in the development and maintenance of prostate tissue. Treatment of prostate cancer by androgen ablation either surgically or biochemically results in massive cell death and tumor regression. However, this is often followed by the onset of aggressive disease, which is fatal. Various studies have been conducted to understand the mechanism leading to the establishment of aggressive disease following treatment. An interesting comprehensive study recently conducted by Chen et al shows the increase in androgen receptor (AR) transcript to be the key factor in disease recurrence following treatment. This up-regulation in the AR levels is shown to increase sensitivity to low levels of androgen, leading to ligand-dependent downstream gene expression and tumor recurrence. A "mass action" model has been proposed to explain this phenomenon. Moreover, the increase in mRNA levels has been shown to facilitate conversion of classic antagonists to agonists of hormones action by the recruitment of a subset of co-activators to the promoters of AR-responsive genes. This study underscores the importance of ARs in the establishment of prostate cancer and offers several insights into the mechanism by which tumors recur following androgen ablation. The study also prompts several questions about the reason behind the observed up-regulation and also the mechanism by which classic antagonists are rendered agonistic. The need for the design of novel therapeutic analogues is also emphasized.
Collapse
Affiliation(s)
- Sunita R Setlur
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
42
|
Abstract
The TGF-beta superfamily is the most versatile considering the ability of its members to regulate proliferation, growth arrest, differentiation, and apoptosis of prostatic stromal and epithelial cells as well as the formation of osteoblastic metastases. TGF-beta mediated action in prostate cells follows a complex signaling pathway from binding and phosphorylation of receptor type II to the TbetaRI kinase to Smad activation, resulting in ligand-induced transcription. TGF-beta as an indirect tumor suppressor, its role of regulating tumor induction, as well as tumor suppression depending on the tissue microenvironment merits further exploration. The rationale for targeting growth factors and their receptors for therapeutic intervention is based upon the fact that these proteins represent the most proximate component of the signal transduction cascade. The alternate targeting of intracellular effectors in the signal transduction may be thwarted by cross talk between signaling pathways (such as the Smads in a dynamic interplay with the androgen receptor). TGF-beta within the context of its well-documented apoptosis regulatory actions in the prostate and the significance its key receptor TbetaRII as a potential tumor suppressor, provides a highly attractive candidate for such targeting with high clinical significance for the treatment and diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Brian Zhu
- Division of Urology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
43
|
Danielpour D. Functions and regulation of transforming growth factor-beta (TGF-β) in the prostate. Eur J Cancer 2005; 41:846-57. [PMID: 15808954 DOI: 10.1016/j.ejca.2004.12.027] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2004] [Revised: 10/24/2004] [Accepted: 12/02/2004] [Indexed: 12/15/2022]
Abstract
The prostate is a highly androgen-dependent tissue that in humans exhibits marked susceptibility to carcinogenesis. The malignant epithelium generated from this tissue ultimately loses dependence on androgens despite retention or amplification of the androgen receptor. Accumulating evidence support that transforming growth factor-beta (TGF-beta) plays key roles in the control of androgen dependence and acquisition of resistance to such hormonal control. Although TGF-beta functions as a key tumour suppressor of the prostate, it can also promote malignant progression and metastasis of the advanced disease, through undefined mechanisms. In addition to giving an overview of the TGF-beta field as related to its function in prostate cancer, this Review focuses on novel findings that support the tumour suppressor function of TGF-beta is lost or altered by changes in the activity of the androgen receptor, insulin-like growth factor-I, Akt, and mTOR during malignant progression. Understanding the mechanisms of cross-talk between TGF-beta and such growth modulators has important implications for the rational therapeutics of prostate cancer.
Collapse
Affiliation(s)
- David Danielpour
- Case Comprehensive Cancer Center and Department of Pharmacology, Case Western Reserve University, Wolstein Research Building, Room 3-532, 2103 Cornell Road, Cleveland, OH 44106, USA.
| |
Collapse
|
44
|
Wang H, Song K, Sponseller TL, Danielpour D. Novel Function of Androgen Receptor-associated Protein 55/Hic-5 as a Negative Regulator of Smad3 Signaling. J Biol Chem 2005; 280:5154-62. [PMID: 15561701 DOI: 10.1074/jbc.m411575200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Androgen receptor-associated protein 55 (ARA55/Hic-5) belongs to the LIM protein superfamily and is featured by three or four N-terminal LD motifs and four C-terminal zinc finger-like LIM domains. Both LD motifs and LIM domains can serve as protein-protein interaction interfaces. Recently, we found that enforced expression of ARA55 inhibits transforming growth factor-beta-mediated up-regulation of Smad binding element-luciferase reporter activity in NRP-154 and NRP-152 rat prostate and LNCaP human prostate cell lines. Moreover, ARA55 also inhibits the induction of Smad-binding element 4-luciferase and 3TP-luciferase (a plasminogen activator inhibitor-1 (PAI-1) promoter construct) reporters by constitutively active (CA)-Smad3 in these cell lines. Co-immunoprecipitation studies suggest an interaction between ARA55 and either CA-Smad3 or wild-type Smad3 in HEK293 cells that occurs through the MH2 domain of Smad3 and the C terminus of ARA55 with wild-type Smad3 having stronger affinity than CA-Smad3 to ARA55. Glutathione S-transferase pull-down assays demonstrate that this interaction can occur in a cell-free system. These results are consistent with the luciferase data showing that the C terminus of ARA55 is critical for suppression of Smad3 activity. Furthermore, using a mammalian two-hybrid system, we confirmed that ARA55 interacts with the MH2 domain of Smad3 and suppresses CA-Smad3-induced transcriptional responses. In conclusion, these results support that ARA55 selectively intercepts transforming growth factor-beta signaling through an interaction of the LIM domain of ARA55 with the MH2 domain of Smad3.
Collapse
Affiliation(s)
- Hui Wang
- Ireland Cancer Center Research Laboratories and Department of Pharmacology, Case Western Reserve University/University Hospitals, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
45
|
Tahmatzopoulos A, Rowland RG, Kyprianou N. The role of alpha-blockers in the management of prostate cancer. Expert Opin Pharmacother 2004; 5:1279-85. [PMID: 15163273 PMCID: PMC2274914 DOI: 10.1517/14656566.5.6.1279] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Prostate cancer is the second most common cause of cancer death in men in the US. Patients with prostate cancer are initially treated with surgical resection, radiation or antiandrogen therapy. After an initial remission, however, the majority of prostate tumours evolve into a highly aggressive, metastatic androgen-independent state, for which successful therapy has not yet been established. During the past few years, new perspectives have emerged towards the development of preventive and therapeutic approaches for prostate cancer. Quinazoline-based alpha(1)-blockers have been shown to have antitumour efficacy against prostate cancer cells in inducing apoptosis and anoikis via an alpha(1)-adrenoceptor-independent mechanism. Specifically, doxazosin and terazosin can induce apoptosis, inhibit invasion and migration of prostate cancer and endothelial cells, and reduce their adhesion potential to extracellular matrix components, thus enhancing their susceptibility to anoikis. This review discusses recent evidence suggesting the apoptotic efficacy of quinazoline-based alpha(1)-adrenoceptor antagonists, doxazosin and terazosin and speculates on the therapeutic promise of these drugs as novel antitumour agents against prostate cancer. From a drug discovery perspective, separation of the effect of doxazosin on apoptosis in prostate cancer cells from its original pharmacological activity in normal prostate cells, will provide a molecular basis in developing a novel class of apoptosis-inducing agents through lead optimisation.
Collapse
|
46
|
Uzgare AR, Xu Y, Isaacs JT. In vitro culturing and characteristics of transit amplifying epithelial cells from human prostate tissue. J Cell Biochem 2004; 91:196-205. [PMID: 14689591 DOI: 10.1002/jcb.10764] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The prostatic epithelium is functionally organized in stem cell units. This unit consists of a slow turn over stem cell within the basal epithelial layer which can replenish itself and provide progeny which differentiate down either a neuroendocrine or exocrine pathway. The maturation along the exocrine pathway initially involves transit amplifying cells within the basal layer proliferating and subsequently the progeny maturing into intermediate cells. These intermediate cells migrate into the luminal layer where they terminally differentiate into non-proliferative secretory luminal cells which express prostate specific differentiation markers, like PSA. A growing body of experimental evidence has identified the proliferating transit amplifying/intermediate cells as the cells of origin for the common prostatic adenocarcinomas. Using a series of growth characteristics, and mRNA and protein markers, we have validated that primary cultures can be established in serum free defined media from surgically resected human prostates which are composed of essentially pure population of transit amplifying cells. At each serial passage, the subsequent cultures undergo enhanced maturation into intermediate cells and by the 7-10th passage these cells eventually lose their proliferative ability. This study validates that these cells are a useful and relevant system for the determination of molecular events involved in prostatic carcinogenesis.
Collapse
Affiliation(s)
- Aarti R Uzgare
- Division of Experimental Therapeutics, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA
| | | | | |
Collapse
|
47
|
Abstract
RUNX3: is expressed by gastric epithelial cells throughout development. Mice whose Runx3 gene has been knocked out died soon after birth. In the knockout mouse, gastric epithelia exhibited hyperplasia and epithelial apoptosis was suppressed. Analysis using a primary culture system for the epithelial cells suggested that this is caused by the reduced sensitivity of Runx3-/- gastric epithelial cells to the growth-inhibiting and apoptosis-inducing activities of TGF-beta. In human and mouse gastric cancer cell lines, RUNX3/Runx3 was silenced due to hypermethylation of CpG islands in the promoter region. Exogenous expression of RUNX3 in the cells that do not express the endogenous gene caused an inhibition of growth both in vivo and in vitro. These observations indicate that Runx3 is a major growth regulator of gastric epithelial cells, and that it is deeply involved in gastric tumorigenesis in both humans and mice.
Collapse
Affiliation(s)
- Hiroshi Fukamachi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | |
Collapse
|
48
|
Abstract
The normal development and maintenance of the prostate is dependent on androgen acting through the androgen receptor (AR). AR remains important in the development and progression of prostate cancer. AR expression is maintained throughout prostate cancer progression, and the majority of androgen-independent or hormone refractory prostate cancers express AR. Mutation of AR, especially mutations that result in a relaxation of AR ligand specificity, may contribute to the progression of prostate cancer and the failure of endocrine therapy by allowing AR transcriptional activation in response to antiandrogens or other endogenous hormones. Similarly, alterations in the relative expression of AR coregulators have been found to occur with prostate cancer progression and may contribute to differences in AR ligand specificity or transcriptional activity. Prostate cancer progression is also associated with increased growth factor production and an altered response to growth factors by prostate cancer cells. The kinase signal transduction cascades initiated by mitogenic growth factors modulate the transcriptional activity of AR and the interaction between AR and AR coactivators. The inhibition of AR activity through mechanisms in addition to androgen ablation, such as modulation of signal transduction pathways, may delay prostate cancer progression.
Collapse
Affiliation(s)
- Cynthia A Heinlein
- George Whipple Laboratory for Cancer Research, Department of Pathology, University of Rochester, Rochester, NY 14642, USA
| | | |
Collapse
|
49
|
Affiliation(s)
- Peter M Siegel
- Cancer Biology and Genetics Program, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | |
Collapse
|
50
|
Berger AP, Kofler K, Bektic J, Rogatsch H, Steiner H, Bartsch G, Klocker H. Increased growth factor production in a human prostatic stromal cell culture model caused by hypoxia. Prostate 2003; 57:57-65. [PMID: 12886524 DOI: 10.1002/pros.10279] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Local hypoxia may be one of the triggers of embryonic reawakening of the stroma and subsequent hyperplastic growth in the prostate. Using a cell culture model of human prostatic stromal cells, we investigated the effects of hypoxia on activation of hypoxia-inducible factor 1 (HIF 1) and on the production of growth factors. METHODS Primary prostatic stromal cells were grown in normal and hypoxic (1% O(2)) atmosphere. Activation of HIF 1 was evaluated after different time intervals by Western blot. Induced secretion of growth factors VEGF, FGF-7, TGF-beta, IL 8, and FGF-2 were analyzed by ELISA. To confirm the in vitro findings we also performed immunohistochemistry of HIF 1alpha as well as pro-collagen I, collagens I, III, and IV in the benign tissue of radical prostatectomy specimens. RESULTS HIF 1 is activated in a time-dependent manner, already starting 1 hr after exposure of stromal cells to hypoxic conditions. Secretion of VEGF, FGF-7, TGF-beta, FGF-2, and IL 8 is increased under hypoxic in vitro conditions in comparison to normoxia. Levels of TGF-beta, VEGF, and IL 8 were rapidly and statistically significantly increased in the supernatant of hypoxic cells. Consistent with the in vitro findings, immunohistochemistry of HIF 1alpha in (benign prostatic hyperplasia) BPH tissue revealed strong HIF 1alpha nuclear staining in hyperplastic areas. No difference was observed in the collagen pattern between hyperplastic and normal prostate tissue. CONCLUSIONS Prostatic stromal cells respond to hypoxia by upregulation of secretion of several growth factors suggesting that hypoxia can trigger prostatic growth. Therefore, hypoxia might be a key factor contributing to the pathogenesis of BPH.
Collapse
|