1
|
Kasai K, Kato T, Kadota Y, Erdenebayar O, Keyama K, Kawakita T, Yoshida K, Kuwahara A, Matsuzaki T, Irahara M. Intraperitoneal administration of activin A promotes development of endometriotic lesions in a mouse model of endometriosis. THE JOURNAL OF MEDICAL INVESTIGATION 2019; 66:123-127. [PMID: 31064924 DOI: 10.2152/jmi.66.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
PURPOSE This study aimed to investigate the effect of intraperitoneal administration of activin on the occurrence of endometriosis using a mouse model of endometriosis. METHODS A mouse model of endometriosis was prepared by intraperitoneally administering endometrial tissue and blood collected from donor mice to C57BL/6J 7-8- week-old recipient mice. A total of 400 μg of activin A was intraperitoneally administered to model mice in the activin group for 5 days. Intraperitoneal endometriotic lesions were confirmed macroscopically and IL-6 and TNF-α levels in washed ascites were measured by ELISA. RESULTS Endometriotic lesions were observed in all mice. In the activin group, the maximum diameter of endometriotic lesions was significantly larger than that in control group (4.7?1.3 vs 2.9?0.9 mm, p?0.01). The total area of the lesion was also significantly higher in the activin group than in the control group (21.1?9.9 vs 8.8?5.4 mm2,p?0.01). Furthermore, IL-6 and TNF-α levels in ascites were significantly higher in the activin group than in the control group (IL-6 : 85.8?15.3 vs 75.1?19.3 pg/ml, p?0.05 ; TNF-α : 629.8?15.4 vs 605.9?11.4 pg/ml, p?0.05). CONCLUSION Activin promotes occurrence of endometriosis. Inflammatory cytokines are also elevated by activin administration,suggesting that they may contribute to progression of endometriosis J. Med. Invest. 66 : 123-127, February, 2019.
Collapse
Affiliation(s)
- Kana Kasai
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Kato
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuri Kadota
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Otgontsetseg Erdenebayar
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kaoru Keyama
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takako Kawakita
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kanako Yoshida
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akira Kuwahara
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Toshiya Matsuzaki
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Minoru Irahara
- Department of Obstetrics and Gynecology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
2
|
Miles FL, Kurtoglu S, Ahmer C, Soori M, Favate JS, Sikes RA. Transforming growth factor-β signaling induced during prostate cancer cell death and neuroendocrine differentiation is mediated by bone marrow stromal cells. Prostate 2015; 75:1802-13. [PMID: 26392321 DOI: 10.1002/pros.23060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/22/2015] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Prostate cancer that has metastasized to bone undergoes critical interactions with bone marrow stromal cells (BMSCs), ultimately promoting tumor survival. Previous studies have shown that BMSCs secrete factors that promote prostate cancer apoptosis or neuroendocrine differentiation. Because of the significance of transforming growth factor-β (TGF-β) family cytokines in cytostasis and bone metastasis, the role of TGF-β signaling in the context of prostate cancer-BMSC interactions was investigated. METHODS The role of TGF-β family signaling in BMSC-induced apoptosis of lineage-related prostate cancer cells was investigated in live/dead assays. SMAD phosphorylation or activity during apoptosis and neuroendocrine differentiation was investigated using immunofluorescence, Western blotting, and luciferase reporter assays, along with the ALK-4, -5, -7 kinase inhibitor, SB-431542. RESULTS Treatment of castration-resistant prostate cancer cells with SB-431542 resulted in significant reduction of apoptosis mediated by HS-5 BMSCs, supporting the involvement of TGF-β/SMAD signaling during this event. Interestingly, however, pre-treatment of BMSCs with TGF-β1 (5 ng/mL) yielded a conditioned medium that elicited a marked reduction in prostate cancer death. Phosphorylated-SMAD2 (P-SMAD2) was activated in BMSC-triggered transdifferentiated prostate cancer cells, as demonstrated through immunoblotting and luciferase reporter assays. However, SB-431542 did not restore androgen receptor and prostate specific antigen levels down-regulated by BMSC-secreted factors. Prostate cancer cells induced to undergo neuroendocrine differentiation in a BMSC-independent mechanism also showed elevated levels of P-SMAD2. DISCUSSION Collectively, our findings indicate that: (1) TGF-β family cytokines or regulated factors secreted from BMSCs are involved in prostate cancer apoptosis; (2) TGF-β signaling in prostate cancer cells is induced during neuroendocrine differentiation; and (3) TGF-β1 stimulation of BMSCs alters paracrine signaling to create a permissive environment for prostate cancer survival, suggesting a mechanism for prostate cancer-mediated colonization of bone. CONCLUSIONS TGF-β signaling resulting in activation of SMAD2 in prostate cancer may be an indicator of cellular stress in the presence of toxic paracrine factors released from the bone marrow stroma, ultimately fostering prostate cancer colonization of bone.
Collapse
Affiliation(s)
- Fayth L Miles
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
- Department of Biological Sciences, Center for Translational Cancer Research, University of Delaware, Newark, Delaware
- Department of Epidemiology, Fielding School of Public Health, University of California-Los Angeles, Los Angeles, California
| | - Senem Kurtoglu
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
- Department of Biological Sciences, Center for Translational Cancer Research, University of Delaware, Newark, Delaware
| | - Chris Ahmer
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
| | - Mehrnoosh Soori
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
| | - John S Favate
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
| | - Robert A Sikes
- Laboratory for Cancer Ontogeny and Therapeutics, University of Delaware, Newark, Delaware
- Department of Biological Sciences, Center for Translational Cancer Research, University of Delaware, Newark, Delaware
| |
Collapse
|
3
|
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
|
4
|
Hofland J, van Weerden WM, Steenbergen J, Dits NFJ, Jenster G, de Jong FH. Activin A stimulates AKR1C3 expression and growth in human prostate cancer. Endocrinology 2012; 153:5726-34. [PMID: 23024260 DOI: 10.1210/en.2011-2065] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Local androgen synthesis in prostate cancer (PC) may contribute to the development of castration-resistant PC (CRPC), but pathways controlling intratumoral steroidogenic enzyme expression in PC are unknown. We investigated the effects of activin, a factor involved in the regulation of PC growth and steroidogenic enzyme expression in other steroidogenic tissues, on intratumoral steroidogenesis in PC. Activin A effects and regulation of the activin-signaling pathway molecules were studied in the PC cell lines LNCaP, VCaP, and PC-3 and in 13 individual PC xenograft models. Also, expression levels of inhibin βA- and βB-subunits (INHBA and INHBB) and of the activin antagonist follistatin were quantitated in patient PC tissues. Activin A induced the expression and enzyme activity of 17β-hydroxysteroid dehydrogenase enzyme AKR1C3 in LNCaP and VCaP cells. Inhibition of endogenous activin A action in the PC-3 cell line decreased AKR1C3 levels and consequently testosterone synthesis. In return, androgens suppressed INHBA expression in both VCaP cells and the PC xenograft models. The antiproliferative effects of activin A were opposed by physiological concentrations of androstenedione in LNCaP cells. In patient PC tissues, expression levels of INHBA were increased in CRPC samples and correlated with AKR1C3 levels. Moreover, a high ratio of activin subunits to follistatin was associated with a worse metastasis-free survival in patients. In conclusion, activin A is controlled by androgens in PC models and regulates local androgen production. Activin A thus seems to mediate (residual) intratumoral androgen levels and could form a novel therapeutic target in CRPC.
Collapse
Affiliation(s)
- Johannes Hofland
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
5
|
Abstract
Activins are the members of transforming growth factor β superfamily and act as secreted proteins; they were originally identified with a reproductive function, acting as endocrine-derived regulators of pituitary follicular stimulating hormone. In recent years, additional functions of activins have been discovered, including a regulatory role during crucial phases of growth, differentiation, and development such as wound healing, tissue repair, and regulation of branching morphogenesis. The functions of activins through activin receptors are pleiotrophic, while involving in the etiology and pathogenesis of a variety of diseases and being cell type-specific, they have been identified as important players in cancer metastasis, immune responses, inflammation, and are most likely involved in cell migration. In this chapter, we highlight the current knowledge of activin signaling and discuss the potential physiological and pathological roles of activins acting on the migration of various cell types.
Collapse
|
6
|
|
7
|
Kang HY, Huang HY, Hsieh CY, Li CF, Shyr CR, Tsai MY, Chang C, Chuang YC, Huang KE. Activin A enhances prostate cancer cell migration through activation of androgen receptor and is overexpressed in metastatic prostate cancer. J Bone Miner Res 2009; 24:1180-1193. [PMID: 19257827 DOI: 10.1359/jbmr.090219] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bone metastasis is the major cause of mortality associated with prostate cancer. Whereas activin A is known to inhibit prostate cancer cell growth and promote apoptosis, the correlation of elevated activin A with increasing serum prostate-specific antigen (PSA) levels in bone metastatic stages of prostate cancer is well documented. The molecular mechanisms explaining these paradoxical effects of activin A and how activin A influences the progression of prostate cancer with bone metastasis remain unclear. By comparing expression profiles of primary prostate cancer biopsies, with and without bone metastasis, we discovered that the expression of activin A is increased in cases with bone metastatic propensity and correlates with increased androgen receptor (AR), PSA expression, and Gleason scores. Activin A promotes migration of prostate cancer cells to osteoblasts, elevates the AR gene transcription through Smads through binding to AR promoter, and induces nuclear translocation of AR to interact with Smad3. Knockdown of Smad3 by siRNA decreases activin A-promoted AR expression and cancer cell migration. Overexpression of AR reversed Smad3-siRNA suppression on activin A-mediated cell migration to osteoblasts. These data suggest that activation of the AR through Smads is required for activin A-promoted prostate cancer cell migration to bone matrix, thereby promoting the bone metastatic phenotype, and the activin A-Smad-AR axis may be considered a therapeutic target in bone metastatic diseases.
Collapse
Affiliation(s)
- Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung Memorial Hospital-Koahsiung Medical Center, Chang Gung University, College of Medicine, Kaohsiung, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Gupta V, Bhasin S, Guo W, Singh R, Miki R, Chauhan P, Choong K, Tchkonia T, Lebrasseur NK, Flanagan JN, Hamilton JA, Viereck JC, Narula NS, Kirkland JL, Jasuja R. Effects of dihydrotestosterone on differentiation and proliferation of human mesenchymal stem cells and preadipocytes. Mol Cell Endocrinol 2008; 296:32-40. [PMID: 18801408 PMCID: PMC2873614 DOI: 10.1016/j.mce.2008.08.019] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 07/29/2008] [Accepted: 08/10/2008] [Indexed: 10/21/2022]
Abstract
UNLABELLED The mechanisms by which androgens regulate fat mass are poorly understood. Although testosterone has been reported to increase lipolysis and inhibit lipid uptake, androgen effects on proliferation and differentiation of human mesenchymal stem cells (hMSCs) and preadipocytes have not been studied. Here, we investigated whether dihydrotestosterone (DHT) regulates proliferation, differentiation, or functional maturation of hMSCs and human preadipocytes from different fat depots. DHT (0-30 nM) dose-dependently inhibited lipid accumulation in adipocytes differentiated from hMSCs and downregulated expression of aP2, PPARgamma, leptin, and C/EBPalpha. Bicalutamide attenuated DHT's inhibitory effects on adipogenic differentiation of hMSCs. Adipocytes differentiated in presence of DHT accumulated smaller oil droplets suggesting reduced extent of maturation. DHT decreased the incorporation of labeled fatty acid into triglyceride, and downregulated acetyl CoA carboxylase and DGAT2 expression in adipocytes derived from hMSCs. DHT also inhibited lipid accumulation and downregulated aP2 and C/EBPalpha in human subcutaneous, mesenteric and omental preadipocytes. DHT stimulated forskolin-stimulated lipolysis in subcutaneous and mesenteric preadipocytes and inhibited incorporation of fatty acid into triglyceride in adipocytes differentiated from preadipocytes from all fat depots. CONCLUSIONS DHT inhibits adipogenic differentiation of hMSCs and human preadipocytes through an AR-mediated pathway, but it does not affect the proliferation of either hMSCs or preadipocytes. Androgen effects on fat mass represent the combined effect of decreased differentiation of fat cell precursors, increased lipolysis, and reduced lipid accumulation.
Collapse
Affiliation(s)
- Vandana Gupta
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Shalender Bhasin
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Wen Guo
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Rajan Singh
- Charles R. Drew University, Los Angeles, CA 90059, United States
| | - Rika Miki
- Charles R. Drew University, Los Angeles, CA 90059, United States
| | - Pratibha Chauhan
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Karen Choong
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Tamara Tchkonia
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Nathan K. Lebrasseur
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - John N. Flanagan
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - James A. Hamilton
- Department of Biophysics, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Jason C. Viereck
- Department of Biophysics, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Navjot S. Narula
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - James L. Kirkland
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Ravi Jasuja
- Department of Medicine, Boston University, School of Medicine, Boston Medical Center, Boston, MA 02118, United States
- Charles R. Drew University, Los Angeles, CA 90059, United States
| |
Collapse
|
9
|
Sampson N, Madersbacher S, Berger P. [Pathophysiology and therapy of benign prostatic hyperplasia]. Wien Klin Wochenschr 2008; 120:390-401. [PMID: 18726663 DOI: 10.1007/s00508-008-0986-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 05/21/2008] [Indexed: 12/17/2022]
Abstract
Benign prostatic hyperplasia (BPH) and benign prostatic enlargement (BPE) are among the most frequent medical disorders of elderly men and cause a number of annoying symptoms of the lower urinary tract (LUTS), leading to reduced quality of life and severe complications, including acute urinary retention. Nodular overgrowth of the epithelium and in particular the fibromuscular tissue is observed in the transition zone and periurethral areas. In particular, functional and phenotypic transdifferentiation of fibroblasts into myofibroblasts is a hallmark of the tissue remodeling in the benign hyperplastic prostate. BPH/BPE have a complex pathophysiology with a multitude of endocrine and local factors involved. Two risk factors, namely aging and circulating androgens, contribute significantly to risk of BPH/BPE. One of the primary initiating mechanisms appears to be a consequence of age-related changes in systemic sex steroid hormone levels accompanied by alterations in local androgen metabolism. This results in the disruption of the delicate balance of interacting growth factor signaling pathways and stromal/epithelial interactions generating a growth promoting and tissue remodeling microenvironment that leads to an increase in prostate volume. Secondarily, altered cytokine and chemoattractant production by the remodeled stroma promotes local inflammation that may further contribute to disease progression via lymphocyte-derived inflammatory cytokines and reactive oxygen/nitrogen species. Local hypoxia as a result of increased oxygen demands of proliferating cells may induce low levels of reactive oxygen species promoting neovascularization and fibroblast-to-myofibroblast transdifferentiation. Medical therapies for LUTS due to BPH/BPE have changed little over the past 15 years with mainstay treatments being alpha-adrenoreceptor blockade and 5alpha-reductase inhibitors. We provide an in depth view of the mechanisms underlying BPH/BPE and relate new research findings to the clinical picture with the prospect of novel therapeutic targets, including selective hormone antagonists/agonists, anti-stromal therapy, vitamin-D analogues and approaches to redress the redox imbalance.
Collapse
Affiliation(s)
- Natalie Sampson
- Institut für Biomedizinische Alternsforschung, Osterreichische Akademie der Wissenschaften, Innsbruck, Austria
| | | | | |
Collapse
|
10
|
Shidaifat F, Al-Zuhair I, Bani-Ismail Z. Interaction of testosterone with inhibin alpha and betaA subunits to regulate prostate gland growth. Endocrine 2007; 31:38-43. [PMID: 17709896 DOI: 10.1007/s12020-007-0011-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/01/2022]
Abstract
Testosterone regulation of prostate gland growth has been shown to involve reciprocal interaction with inhibin and activin. This study was therefore conducted to correlate the effect of testosterone on prostate gland proliferation and differentiation with the level of expression of inhibin alpha and betaA subunits. Immature dogs were treated with testosterone for 0, 3, 7, and 14 days and prostate gland growth was assessed by morphological and immunohistological localization of differentiation and proliferation markers. The results showed that testosterone treatment resulted in an initial significant increase in PCNA proliferation index by days 3 and 7, followed by a significant decrease by day 14 post-treatment. Interestingly, the decrease of cell proliferation was associated with structural and biochemical changes characteristic of glandular and stromal differentiation of the prostate gland. These changes include progressive glandular ductal canalization and inter-ductal stroma differentiation which were apparent from a gradual shift from vimentin expression to vimentin and alpha-actin expression. Testosterone also had a differential effect on inhibin alpha and beta subunits. Although testosterone treatment resulted in significant and constant inhibition of alpha subunit mRNA expression, it resulted in a significant increase of betaA mRNA expression by day 3, followed by a decrease by days 7 and 14. These results indicated that testosterone acts first to drive proliferation of undifferentiated prostatic cells and then to maintain a low proliferation turnover of differentiated cells. Because it has been shown that activin is an antagonistic regulator of androgens, the attenuated stimulatory effect of testosterone on cell proliferation by day 14 might be mediated, at least in part, by interplay between testosterone and activin.
Collapse
Affiliation(s)
- Falah Shidaifat
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan.
| | | | | |
Collapse
|
11
|
Yang C, Trent S, Ionescu-Tiba V, Lan L, Shioda T, Sgroi D, Schmidt EV. Identification of cyclin D1- and estrogen-regulated genes contributing to breast carcinogenesis and progression. Cancer Res 2007; 66:11649-58. [PMID: 17178859 DOI: 10.1158/0008-5472.can-06-1645] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumors can become lethal when they progress from preinvasive lesions to invasive carcinomas. Here, we identify candidate tumor progression genes using gene array analysis of preinvasive and invasive tumors from mice, which were then evaluated in human cancers. Immediate early response protein IEX-1, small stress protein 1 (HSPB8), and tumor necrosis factor-associated factor-interacting protein mRNAs displayed higher expression levels in invasive lesions than in preinvasive lesions using samples obtained by laser capture microdissection (LCM) from transgenic erbB2, ras, and cyclin D1 mice. LCM-isolated tissues from patient-matched normal, ductal carcinoma in situ, and invasive ductal carcinoma revealed similar increased expression in invasive human cancers compared with preinvasive and normal samples. These genes induced anchorage independence, increased cell proliferation, and protected against apoptosis, singly or in collaboration with erbB2. Surprisingly, they were all up-regulated by 17beta-estradiol and cyclin D1, and cyclin D1 overexpression increased p300/CBP binding to their promoters, supporting the model that cyclin D1-estrogen receptor (ER) coactivator interactions may be important to its role in ER-positive breast cancer. Additionally, an irreversible dual kinase inhibitor of ErbB signaling inhibited expression of the same genes. The up-regulation of genes contributing to increased invasiveness of ER-positive cancers offers a novel explanation for the contribution of cyclin D1 to a worse prognosis in ER-positive cancers. As targets of estrogen, cyclin D1, and erbB2 signaling, these candidates offer insights into the nature of the second events involved in breast cancer progression, regulatory events contributing to invasion, and potential targets of combined inhibition of hormone and growth factor signaling pathways.
Collapse
Affiliation(s)
- Chuanwei Yang
- Massachusetts General Hospital Cancer Research Center, Massachusetts General Hospital Cancer Center--Harvard University, 55 Fruit Street, GRJ 904, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Ageing of the male reproductive system is characterized by changes in the endocrine system, hypogonadism, erectile dysfunction and proliferative disorders of the prostate gland. Stochastic damage accumulating within ageing leads to progressive dysregulation at each level of the hypothalamic-pituitary-gonadal (HPG) axis and in local auto/paracrine interactions, thereby inducing morphological changes in reproductive target organs, such as the prostate, testis and penis. Despite age-related changes in the HPG axis, endocrine functions are generally sufficient to maintain fertility in elderly men. Ageing of the male reproductive system can give rise to clinically relevant manifestations, such as benign prostatic hyperplasia (BPH), prostate cancer (PCa) and erectile dysfunction (ED). In this review, we discuss morphological/histological changes occurring in these organs and current views and concepts of the underlying pathology. Moreover, we emphasize the molecular/cellular pathways leading to reduced testicular/penile function and proliferative disorders of the prostate gland.
Collapse
Affiliation(s)
- N Sampson
- Institute for Biomedical Ageing Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | |
Collapse
|
13
|
Leto G, Incorvaia L, Badalamenti G, Tumminello FM, Gebbia N, Flandina C, Crescimanno M, Rini G. Activin A circulating levels in patients with bone metastasis from breast or prostate cancer. Clin Exp Metastasis 2006; 23:117-22. [PMID: 16841234 DOI: 10.1007/s10585-006-9010-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Accepted: 04/10/2006] [Indexed: 10/24/2022]
Abstract
Recent studies have highlighted that Activin A, a member of the transforming growth factor-beta (TGF-beta) superfamily, may be involved in the regulation of osteoblastic activity and in osteoclast differentiation. Therefore, we have investigated the clinical significance of its circulating levels in patients with bone metastasis. Activin A serum concentrations were determined, by a commercially available enzyme-linked immunosorbent assay kit, in 72 patients with breast cancer (BC) or prostatic cancer (PC) with (BM+) or without (BM-) bone metastases, in 15 female patients with age-related osteoporosis (OP), in 20 patients with benign prostatic hypertrophy (BPH) and in 48 registered healthy blood donors (HS) of both sex (25 female and 23 male). Activin A serum concentrations were significantly increased in BC or PC patients as compared to OP (P < 0.0001) or BPH (P = 0.045), respectively, or to sex matched HS (P < 0.0001). Additionally, these levels resulted more elevated in PC patients as compared to BC patients (P = 0.032). Interestingly, Activin A was significantly higher in BM+ patients than in BM- patients (BC, P = 0.047; PC, P = 0.016). In BC patients, a significant correlation was observed only between Activin A and number of bone metastases (P = 0.0065) while, in PC patients, Activin A levels were strongly correlated with the Gleason score (P = 0.011) or PSA levels (P = 0.0001) and, to a lessen extent, with the number of bone metastases (P = 0.056). Receiver operating characteristic curve (ROC) analysis showed a fair diagnostic accuracy of Activin A to discriminate between BM+ and BM- patients (BC: AUC = 0.71 +/- 0.09, P = 0.03; PC: AUC = 0.73 +/- 0.081, P = 0.005). These findings indicate that Activin A may be implicated in the pathogenesis of bone metastasis. Therefore, this cytokine may be considered a novel potential target for a more selective therapeutic approach in the treatment of skeletal metastasis and may be also useful as additional biochemical marker of metastatic bone disease.
Collapse
Affiliation(s)
- Gaetano Leto
- Laboratory of Experimental Chemotherapy, Department of Surgery and Oncology, Policlinico Universitario P Giaccone, Via del Vespro 129, 90127, Palermo, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
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: 16] [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
|
15
|
Al-Omari R, Shidaifat F, Dardaka M. Castration induced changes in dog prostate gland associated with diminished activin and activin receptor expression. Life Sci 2005; 77:2752-9. [PMID: 15978633 DOI: 10.1016/j.lfs.2005.03.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Accepted: 03/02/2005] [Indexed: 11/20/2022]
Abstract
This study was conducted to evaluate the effect of androgen ablation on dog prostate gland structure and the proliferation capacity of the prostatic cells and their association with the expression of Activin A and Activin RIIA receptor. The effect of androgen on the prostate gland was compared in intact and castrated dogs after one and two weeks. Specific primary antibodies were used to immunolocalize activin-A, activin receptor type II A and the proliferation marker (PCNA). The results showed that the glandular acini of the prostate gland of intact dogs are lined by tall columnar secretory cells and less abundant flattened basal cells and surrounded by a thin fibromuscular tissue. The cytoplasm of the glandular cells exhibited an intense immunoreaction for activin A and activin RIIA receptor while basal cells expressed PCNA. Castration induced a remarkable atrophy of the prostatic acini associated with a progressive loss of secretory epithelial cells, which showed a dramatic decrease to complete disappearance of Activin A and Activin RIIA receptor immunoreactions. The remaining cells of the atrophied acini continue to express PCNA and the inter-acinar fibromuscular tissue showed a remarkable increase in its mass and are induced to express PCNA. These results indicated that androgen is required for the survival of epithelial cells and to maintain growth-quiescent fibromuscular cells, while basal cell proliferation is androgen independent. The changes in the Activin A and Activin RIIA receptor localization and their association with the dynamic pattern of prostate gland regression after castration suggested that Activin A and Activin RIIA receptor expression are androgen dependent.
Collapse
Affiliation(s)
- Ruba Al-Omari
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid, Jordan
| | | | | |
Collapse
|
16
|
Chen G, Nomura M, Morinaga H, Matsubara E, Okabe T, Goto K, Yanase T, Zheng H, Lu J, Nawata H. Modulation of Androgen Receptor Transactivation by FoxH1. J Biol Chem 2005; 280:36355-63. [PMID: 16120611 DOI: 10.1074/jbc.m506147200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Androgen signaling plays key roles in the development and progression of prostate cancer, and numerous ongoing studies focus on the regulation of androgen receptor (AR) transactivity to develop novel therapies for the treatment of androgen-independent prostate cancer. FoxH1, a member of the Forkhead-box (FOX) gene family of transcription factors, takes part in mediating transforming growth factor-beta/activin signaling through its interaction with the Smad2.Smad4 complex. Using a series of experiments, we found that FoxH1 repressed both ligand-dependent and -independent transactivation of the AR on androgen-induced promoters. This action of FoxH1 was independent of its transactivation capacity and activin A but relieved by Smad2.Smad4. In addition, the repression of the AR by FoxH1 did not require deacetylase activity. A protein-protein interaction was identified between the AR and FoxH1 independently of dihydrotestosterone. Furthermore, a confocal microscopic analysis of LNCaP cells revealed that the interaction between the AR and FoxH1 occurred in the nucleus and that FoxH1 specifically blocked the foci formation of dihydrotestosterone-activated AR, which has been shown to be correlated with the AR transactivation potential. Taken together, our results indicate that FoxH1 functions as a new corepressor of the AR. Our observations not only strengthen the role of FoxH1 in AR-mediated transactivation but also suggest that therapeutic interventions based on AR-coregulator interactions could be designed to block both androgen-dependent and -independent growth of prostate cancer.
Collapse
Affiliation(s)
- Guangchun Chen
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
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: 5.0] [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
|