1
|
Hanamura T, Christenson JL, O'Neill KI, Rosas E, Spoelstra NS, Williams MM, Richer JK. Secreted indicators of androgen receptor activity in breast cancer pre-clinical models. Breast Cancer Res 2021; 23:102. [PMID: 34736512 PMCID: PMC8567567 DOI: 10.1186/s13058-021-01478-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Accumulating evidence has attracted attention to the androgen receptor (AR) as a biomarker and therapeutic target in breast cancer. We hypothesized that AR activity within the tumor has clinical implications and investigated whether androgen responsive serum factors might serve as a minimally invasive indicator of tumor AR activity. METHODS Based on a comprehensive gene expression analysis of an AR-positive, triple negative breast cancer patient-derived xenograft (PDX) model, 163 dihydrotestosterone (DHT)-responsive genes were defined as an androgen responsive gene set. Among them, we focused on genes that were DHT-responsive that encode secreted proteins, namely KLK3, AZGP1 and PIP, that encode the secreted factors prostate specific antigen (PSA), zinc-alpha-2-glycoprotein (ZAG) and prolactin induced protein (PIP), respectively. Using AR-positive breast cancer cell lines representing all breast cancer subtypes, expression of candidate factors was assessed in response to agonist DHT and antagonist enzalutamide. Gene set enrichment analysis (GSEA) was performed on publically available gene expression datasets from breast cancer patients to analyze the relationship between genes encoding the secreted factors and other androgen responsive gene sets in each breast cancer subtype. RESULTS Anti-androgen treatment decreased proliferation in all cell lines tested representing various tumor subtypes. Expression of the secreted factors was regulated by AR activation in the majority of breast cancer cell lines. In GSEA, the candidate genes were positively correlated with an androgen responsive gene set across breast cancer subtypes. CONCLUSION KLK3, AZGP1 and PIP are AR regulated and reflect tumor AR activity. Further investigations are needed to examine the potential efficacy of these factors as serum biomarkers.
Collapse
Affiliation(s)
- Toru Hanamura
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Emmanuel Rosas
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Michelle M Williams
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
2
|
Pietri E, Massa I, Bravaccini S, Ravaioli S, Tumedei MM, Petracci E, Donati C, Schirone A, Piacentini F, Gianni L, Nicolini M, Campadelli E, Gennari A, Saba A, Campi B, Valmorri L, Andreis D, Fabbri F, Amadori D, Rocca A. Phase II Study of Dehydroepiandrosterone in Androgen Receptor-Positive Metastatic Breast Cancer. Oncologist 2019; 24:743-e205. [PMID: 30591548 PMCID: PMC6656524 DOI: 10.1634/theoncologist.2018-0243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/28/2018] [Indexed: 12/02/2022] Open
Abstract
LESSONS LEARNED The androgen receptor (AR) is present in most breast cancers (BC), but its exploitation as a therapeutic target has been limited.This study explored the activity of dehydroepiandrosterone (DHEA), a precursor being transformed into androgens within BC cells, in combination with an aromatase inhibitor (to block DHEA conversion into estrogens), in a two-stage phase II study in patients with AR-positive/estrogen receptor-positive/human epidermal growth receptor 2-negative metastatic BC.Although well tolerated, only 1 of 12 patients obtained a prolonged clinical benefit, and the study was closed after its first stage for poor activity. BACKGROUND Androgen receptors (AR) are expressed in most breast cancers, and AR-agonists have some activity in these neoplasms. We investigated the safety and activity of the androgen precursor dehydroepiandrosterone (DHEA) in combination with an aromatase inhibitor (AI) in patients with AR-positive metastatic breast cancer (MBC). METHODS A two-stage phase II study was conducted in two patient cohorts, one with estrogen receptor (ER)-positive (resistant to AIs) and the other with triple-negative MBC. DHEA 100 mg/day was administered orally. The combination with an AI aimed to prevent the conversion of DHEA into estrogens. The main endpoint was the clinical benefit rate. The triple-negative cohort was closed early. RESULTS Twelve patients with ER-positive MBC were enrolled. DHEA-related adverse events, reported in four patients, included grade 2 fatigue, erythema, and transaminitis, and grade 1 drowsiness and musculoskeletal pain. Clinical benefit was observed in one patient with ER-positive disease whose tumor had AR gene amplification. There was wide inter- and intra-patient variation in serum levels of DHEA and its metabolites. CONCLUSION DHEA showed excellent safety but poor activity in MBC. Although dose and patient selection could be improved, high serum level variability may hamper further DHEA development in this setting.
Collapse
Affiliation(s)
- Elisabetta Pietri
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ilaria Massa
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Ravaioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Maria Maddalena Tumedei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Caterina Donati
- Oncology Pharmacy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessio Schirone
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federico Piacentini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | | | - Mario Nicolini
- Oncology Day Hospital Unit, Cervesi Hospital, Cattolica, Italy
| | | | - Alessandra Gennari
- Medical Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Beatrice Campi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Linda Valmorri
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daniele Andreis
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Andrea Rocca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
3
|
Histopathologic findings in breast surgical specimens from patients undergoing female-to-male gender reassignment surgery. Mod Pathol 2019; 32:346-353. [PMID: 30310177 DOI: 10.1038/s41379-018-0117-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/21/2018] [Accepted: 08/03/2018] [Indexed: 11/08/2022]
Abstract
Breast reduction surgery or mastectomy following administration of androgen therapy is part of the female-to-male gender reassignment process. Details regarding the histopathologic findings in breast tissue from patients undergoing female-to-male gender reassignment surgery are limited. We reviewed hematoxylin and eosin-stained sections of breast tissue from 148 patients who underwent breast reduction surgery or mastectomy as part of the female-to-male gender reassignment process at our institution between January 2014 and May 2017. The spectrum of histologic features in each case was catalogued. The median patient age was 27 years (range 18-60 years). Lobular atrophy was seen to some degree in 73% of cases and was prominent in 42%. A predominantly fibrotic stroma was seen in 45% of cases and areas resembling the fibrous stage of gynecomastia were seen in 41%. Other features included variably ectatic ducts in 96% of cases, cysts in 42%, apocrine metaplasia in 32%, fibroadenomatous change in 27%, usual ductal hyperplasia in 26%, and pseudoangiomatous stromal hyperplasia in 19%. Five cases (3%) demonstrated atypical hyperplasia (atypical ductal hyperplasia in 2; atypical lobular hyperplasia in 2; both atypical ductal hyperplasia and atypical lobular hyperplasia in 1). One case demonstrated high grade ductal carcinoma in situ. No invasive carcinomas were identified. In conclusion, the majority of breast specimens from patients undergoing female-to-male gender reassignment demonstrate at least some degree of lobular atrophy as well as ectatic ducts, fibrous stroma, and areas resembling the fibrous stage of gynecomastia. Only rare cases showed atypical lesions; the clinical significance of these lesions in this setting is uncertain.
Collapse
|
4
|
Takagi K, Miki Y, Ishida T, Sasano H, Suzuki T. The interplay of endocrine therapy, steroid pathways and therapeutic resistance: Importance of androgen in breast carcinoma. Mol Cell Endocrinol 2018; 466:31-37. [PMID: 28918115 DOI: 10.1016/j.mce.2017.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/04/2017] [Accepted: 09/11/2017] [Indexed: 12/27/2022]
Abstract
A great majority of breast carcinomas expresses estrogen receptor (ER) and estrogens have crucial roles in the progress of breast carcinomas. Endocrine therapy targeting ER and/or intratumoral estrogen production significantly improved clinical outcomes of the patients with ER-positive breast carcinomas. However, resistance to endocrine therapy is often observed and significant number of patients will recur after the treatment. In addition, treatment for the patients with triple-negative breast carcinomas (negative for all ER, progesterone receptor (PR) and HER2) are limited to cytotoxic chemotherapy and novel therapeutic targets need to be identified. In breast carcinoma tissues, not only ER but androgen receptor (AR) is frequently expressed, suggesting pivotal roles of androgens in the progress of breast carcinomas. Growing interest on androgen action as possible therapeutic target has been taken, but androgen action seems quite complicated in breast carcinomas and inconsistent findings has been also proposed. In this review, we will summarize recent studies regarding intratumoral androgen production and its regulation as well as distinct subset of breast carcinomas characterized by activated AR signaling and recent clinical trial targeting AR in the patients with either ER-positive and ER-negative breast carcinomas.
Collapse
Affiliation(s)
- Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
5
|
Lee SR, Park MY, Yang H, Lee GS, An BS, Park BK, Jeung EB, Hong EJ. 5α-dihydrotestosterone reduces renal Cyp24a1 expression via suppression of progesterone receptor. J Mol Endocrinol 2018; 60:159-170. [PMID: 29382742 DOI: 10.1530/jme-17-0187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 12/22/2017] [Indexed: 01/03/2023]
Abstract
Androgens act in concert with vitamin D to influence reabsorption of calcium. However, it is unclear whether androgens directly regulate vitamin D homeostasis or control other cellular events that are related to vitamin D metabolism. To examine whether the expression of vitamin D-related genes in mouse kidney is driven by androgens or androgen-dependent effects, the androgen receptor and other sex steroid receptors were monitored in orchidectomized mice treated with 5α-dihydrotestosterone (DHT). Our results revealed that exposing orchidectomized mice to DHT inhibited the expression of progesterone receptor (Pgr) with or without estrogen receptor α expression, the latter was confirmed by ER-positive (MCF7 and T47D) or -negative (PCT) cells analysis. The loss of Pgr in turn decreased the expression of renal 24-hydroxylase via transcriptional regulation because Cyp24a1 gene has a progesterone receptor-binding site on promoter. When male kidneys preferentially hydroxylate 25-hydroxyvitamin D3 using 24-hydroxylase rather than 25-hydroxyvitamin D3-1-alpha hydroxylase, DHT suppressed the Pgr-mediated 24-hydroxylase expression, and it is important to note that DHT increased the blood 25-hydroxyvitamin D3 levels. These findings uncover an important link between androgens and vitamin D homeostasis and suggest that therapeutic modulation of Pgr may be used to treat vitamin D deficiency and related disorders.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Mi-Young Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Yang
- Korean Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Republic of Korea
| | - Bae-Kuen Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eui-Bae Jeung
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
6
|
Yu Z, He S, Wang D, Patel HK, Miller CP, Brown JL, Hattersley G, Saeh JC. Selective Androgen Receptor Modulator RAD140 Inhibits the Growth of Androgen/Estrogen Receptor-Positive Breast Cancer Models with a Distinct Mechanism of Action. Clin Cancer Res 2017; 23:7608-7620. [PMID: 28974548 DOI: 10.1158/1078-0432.ccr-17-0670] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/14/2017] [Accepted: 09/28/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Steroidal androgens suppress androgen receptor and estrogen receptor positive (AR/ER+) breast cancer cells and were used to treat breast cancer, eliciting favorable response. The current study evaluates the activity and efficacy of the oral selective AR modulator RAD140 in in vivo and in vitro models of AR/ER+ breast cancer.Experimental Design: A series of in vitro assays were used to determine the affinity of RAD140 to 4 nuclear receptors and evaluate its tissue-selective AR activity. The efficacy and pharmacodynamics of RAD140 as monotherapy or in combination with palbociclib were evaluated in AR/ER+ breast cancer xenograft models.Results: RAD140 bound AR with high affinity and specificity and activated AR in breast cancer but not prostate cancer cells. Oral administration of RAD140 substantially inhibited the growth of AR/ER+ breast cancer patient-derived xenografts (PDX). Activation of AR and suppression of ER pathway, including the ESR1 gene, were seen with RAD140 treatment. Coadministration of RAD140 and palbociclib showed improved efficacy in the AR/ER+ PDX models. In line with efficacy, a subset of AR-repressed genes associated with DNA replication was suppressed with RAD140 treatment, an effect apparently enhanced by concurrent administration of palbociclib.Conclusions: RAD140 is a potent AR agonist in breast cancer cells with a distinct mechanism of action, including the AR-mediated repression of ESR1 It inhibits the growth of multiple AR/ER+ breast cancer PDX models as a single agent, and in combination with palbociclib. The preclinical data presented here support further clinical investigation of RAD140 in AR/ER+ breast cancer patients. Clin Cancer Res; 23(24); 7608-20. ©2017 AACR.
Collapse
Affiliation(s)
- Ziyang Yu
- Radius Health, Inc., Waltham, Massachusetts.
| | - Suqin He
- Radius Health, Inc., Waltham, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
7
|
Overcoming aromatase inhibitor resistance in breast cancer: possible mechanisms and clinical applications. Breast Cancer 2017; 25:379-391. [PMID: 28389808 DOI: 10.1007/s12282-017-0772-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
Abstract
Estrogen plays crucial roles in the progression of hormone-dependent breast cancers through activation of nuclear estrogen receptor α (ER). Estrogen is produced locally from circulating inactive steroids and adrenal androgens in postmenopausal women. However, conversion by aromatase is a rate-limiting step in intratumoral estrogen production in breast cancer. Aromatase inhibitors (AIs) inhibit the growth of hormone-dependent breast cancers by blocking the conversion of adrenal androgens to estrogen and by unmasking the inhibitory effect of androgens, acting via the androgen receptor (AR). AIs are thus a standard treatment option for postmenopausal hormone-dependent breast cancer. However, although initial use of AIs provides substantial clinical benefit, some breast cancer patients relapse because of the acquisition of AI resistance. A better understanding of the mechanisms of AI resistance may contribute to the development of new therapeutic strategies and aid in the search for new therapeutic targets and agents. We have investigated AI-resistance mechanisms and established six AI-resistant cell lines. Some of them exhibit estrogen depletion-resistance properties via constitutive ER-activation or ER-independent growth signaling. We examined how breast cancer cells can adapt to estrogen depletion and androgen superabundance. Estrogen and estrogenic androgen produced independently from aromatase contributed to cell proliferation in some of these cell lines, while another showed AR-dependent cell proliferation. Based on these findings, currently proposed AI-resistance mechanisms include an aromatase-independent estrogen-producing pathway, estrogen-independent ER function, and ER-independent growth signaling. This review summarizes several hypotheses of AI-resistance mechanisms and discusses how existing or novel therapeutic agents may be applied to treat AI-resistant breast cancers.
Collapse
|
8
|
Lim E, Tarulli G, Portman N, Hickey TE, Tilley WD, Palmieri C. Pushing estrogen receptor around in breast cancer. Endocr Relat Cancer 2016; 23:T227-T241. [PMID: 27729416 DOI: 10.1530/erc-16-0427] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022]
Abstract
The estrogen receptor-α (herein called ER) is a nuclear sex steroid receptor (SSR) that is expressed in approximately 75% of breast cancers. Therapies that modulate ER action have substantially improved the survival of patients with ER-positive breast cancer, but resistance to treatment still remains a major clinical problem. Treating resistant breast cancer requires co-targeting of ER and alternate signalling pathways that contribute to resistance to improve the efficacy and benefit of currently available treatments. Emerging data have shown that other SSRs may regulate the sites at which ER binds to DNA in ways that can powerfully suppress the oncogenic activity of ER in breast cancer. This includes the progesterone receptor (PR) that was recently shown to reprogram the ER DNA binding landscape towards genes associated with a favourable outcome. Another attractive candidate is the androgen receptor (AR), which is expressed in the majority of breast cancers and inhibits growth of the normal breast and ER-positive tumours when activated by ligand. These findings have led to the initiation of breast cancer clinical trials evaluating therapies that selectively harness the ability of SSRs to 'push' ER towards anti-tumorigenic activity. Our review will focus on the established and emerging clinical evidence for activating PR or AR in ER-positive breast cancer to inhibit the tumour growth-promoting functions of ER.
Collapse
Affiliation(s)
- Elgene Lim
- Garvan Institute of Medical Research and St Vincent's HospitalUniversity of New South Wales, Sydney, New South Wales, Australia
| | - Gerard Tarulli
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Neil Portman
- Garvan Institute of Medical Research and St Vincent's HospitalUniversity of New South Wales, Sydney, New South Wales, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories and Adelaide Prostate Cancer Research CentreUniversity of Adelaide, Adelaide, South Australia, Australia
| | - Carlo Palmieri
- Institute of Translational MedicineUniversity of Liverpool, Clatterbridge Cancer Centre, NHS Foundation Trust, and Royal Liverpool University Hospital, Liverpool, Merseyside, UK
| |
Collapse
|
9
|
Chou FP, Tsai CT, Chiou YS, Chen YJ, Li ME, Guo TW, Lyu JW, Chou SH, Wu TK. An enzymatic approach to configurationally raretrans-androsteronyl-α-glucoside and Its potential anticancer application. Chem Biol Drug Des 2016; 89:61-66. [DOI: 10.1111/cbdd.12830] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/08/2016] [Accepted: 07/04/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Feng-Pai Chou
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Chia-Tse Tsai
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Ya-Sheng Chiou
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Yi-Ju Chen
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Meng-Erh Li
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Ting-Wei Guo
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Jason WenJay Lyu
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Sheng-Hao Chou
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| | - Tung-Kung Wu
- Department of Biological Science and Technology; National Chiao Tung University; Hsin-Chu Taiwan, R.O.C
| |
Collapse
|
10
|
Tan HE, Lake F. Interstitial pneumonitis secondary to leuprorelin acetate for prostate cancer. Respirol Case Rep 2016; 4:e00146. [PMID: 27081487 PMCID: PMC4818587 DOI: 10.1002/rcr2.146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/02/2015] [Accepted: 01/20/2016] [Indexed: 11/17/2022] Open
Abstract
Androgen blockade is standard treatment for advanced prostate cancer. We report an uncommon case of interstitial pneumonitis induced by leuprorelin acetate.
Collapse
Affiliation(s)
- Hsern Ern Tan
- Sir Charles Gairdner Hospital Department of Respiratory Medicine Perth Australia; University of Western Australia School of Medicine and Pharmacology Perth Australia
| | - Fiona Lake
- Sir Charles Gairdner Hospital Department of Respiratory Medicine Perth Australia; University of Western Australia School of Medicine and Pharmacology Perth Australia
| |
Collapse
|
11
|
Feng FY, Speers C, Liu M, Jackson WC, Moon D, Rinkinen J, Wilder-Romans K, Jagsi R, Pierce LJ. Targeted radiosensitization with PARP1 inhibition: optimization of therapy and identification of biomarkers of response in breast cancer. Breast Cancer Res Treat 2014; 147:81-94. [PMID: 25104443 DOI: 10.1007/s10549-014-3085-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/26/2014] [Indexed: 01/22/2023]
Abstract
Sustained locoregional control of breast cancer is a significant issue for certain patients. Inhibition of PARP1 is a promising strategy for radiosensitization (RS). We sought to optimize therapy with PARP1 inhibition and radiation (RT) by establishing the most effective treatment schedule, degree of PARP1-mediated RS, and identify early biomarkers predictive of efficacy in breast cancer models. Using clonogenic survival assays, we assessed intrinsic radiosensitivity and RS induced by PARP1 inhibition in breast cancer cell lines. Potential biomarkers of response were evaluated using western blotting, flow cytometry, and immunofluorescence with validation in vivo using tumor xenograft experiments. Across a panel of BC and normal breast epithelial cell lines, the PARP1 inhibitor ABT-888 preferentially radiosensitizes breast cancer (vs. normal) cells with enhancement ratios (EnhR) up to 2.3 independent of intrinsic BC subtype or BRCA mutational status. Concurrent and adjuvant therapy resulted in the highest EnhR of all schedules tested. The degree of RS did not correlate with pretreatment markers of PARP1 activity, DNA damage/repair, or cell cycle distribution. Increases in PARP1 activity 24 h after RT were associated with sensitivity after combination treatment. Findings were confirmed in breast cancer xenograft models. Our study demonstrates that PARP1 inhibition improves the therapeutic index of RT independent of BC subtype or BRCA1 mutational status and that PARP1 activity may serve as a clinically relevant biomarker of response. These studies have led to a clinical trial (TBCRC024) incorporating intratreatment biomarker analyses of PARP1 inhibitors and RT in breast cancer patients.
Collapse
Affiliation(s)
- Felix Y Feng
- Department of Radiation Oncology, University of Michigan, 1500 East Medical Center Drive, UHB2C490-SPC5010, Ann Arbor, MI, 48109-5010, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bianco-Miotto T, Trotta AP, Need EF, Lee AMC, Ochnik AM, Giorgio L, Leach DA, Swinstead EE, O'Loughlin MA, Newman MR, Birrell SN, Butler LM, Harris JM, Buchanan G. Molecular and structural basis of androgen receptor responses to dihydrotestosterone, medroxyprogesterone acetate and Δ(4)-tibolone. Mol Cell Endocrinol 2014; 382:899-908. [PMID: 24239616 DOI: 10.1016/j.mce.2013.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 10/16/2013] [Accepted: 11/06/2013] [Indexed: 01/29/2023]
Abstract
Medroxyprogesterone acetate (MPA) has widely been used in hormone replacement therapy (HRT), and is associated with an increased risk of breast cancer, possibly due to disruption of androgen receptor (AR) signaling. In contrast, the synthetic HRT Tibolone does not increase breast density, and is rapidly metabolized to estrogenic 3α-OH-tibolone and 3β-OH-tibolone, and a delta-4 isomer (Δ(4)-TIB) that has both androgenic and progestagenic properties. Here, we show that 5α-dihydrotestosterone (DHT) and Δ(4)-TIB, but not MPA, stabilize AR protein levels, initiate specific AR intramolecular interactions critical for AR transcriptional regulation, and increase proliferation of AR positive MDA-MB-453 breast cancer cells. Structural modeling and molecular dynamic simulation indicate that Δ(4)-TIB induces a more stable AR structure than does DHT, and MPA a less stable one. Microarray expression analyses confirms that the molecular actions of Δ(4)-TIB more closely resembles DHT in breast cancer cells than either ligand does to MPA.
Collapse
Affiliation(s)
- Tina Bianco-Miotto
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia
| | - Andrew P Trotta
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia; Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Eleanor F Need
- Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Alice M C Lee
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia
| | - Aleksandra M Ochnik
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia
| | - Lauren Giorgio
- Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Damien A Leach
- Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Erin E Swinstead
- Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Melissa A O'Loughlin
- Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia
| | - Michelle R Newman
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia
| | - Stephen N Birrell
- Department of Surgery, Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia
| | - Jonathan M Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| | - Grant Buchanan
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide and Hanson Institute, Australia; Cancer Biology Group, Basil Hetzel Institute for Translational Health Research, The University of Adelaide, Australia.
| |
Collapse
|
13
|
Need EF, Selth LA, Harris TJ, Birrell SN, Tilley WD, Buchanan G. Research resource: interplay between the genomic and transcriptional networks of androgen receptor and estrogen receptor α in luminal breast cancer cells. Mol Endocrinol 2012; 26:1941-52. [PMID: 23023562 DOI: 10.1210/me.2011-1314] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The cellular response to circulating sex steroids is more than the sum of individual hormone actions, instead representing an interplay between activities of the evolutionarily related steroid hormone receptors. An example of this interaction is in breast cancer, where the risk of dying from estrogen receptor-α (ERα)-positive disease decreases approximately 4-fold when androgen receptor (AR) expression is high. In this study, we used chromatin immunoprecipitation sequencing (ChIP-seq) and microarray expression profiling to investigate the genomic and transcriptional cross talk between AR and ERα signaling in a luminal breast cancer cell line model, ZR-75-1. Expression profiling demonstrated reciprocal interference between 5α-dihydrotestosterone (DHT)- and 17β-estradiol (E(2))-induced transcriptional programs. Specifically, regulation of 26% of E(2) and 15% of DHT target genes was significantly affected by cotreatment with the other hormone, in the majority of cases (78-83%) antagonistically. Pathway analysis suggested that DHT cotreatment, for example, depleted E(2)-regulated pathways in cell survival and proliferation. ChIP-seq identified substantial overlap between the steroid receptor cistromes in ZR-75-1 cells, with 10-13% of AR- and ERα-binding sites located within 10 kb of the other receptor. Enrichment of androgen response elements in ERα-binding sites and vice versa was revealed by motif analysis, and AR-binding sites were enriched about E(2)-responsive genes affected by DHT cotreatment. Targeted ChIP and expression analysis revealed locus-specific outcomes when AR and ERα bind to the same DNA region. This work provides the first cistrome data for two steroid receptors in the same cell, insight into the antagonistic interplay between estrogens and androgens in luminal breast cancer, and an important resource for future work aimed at evaluating interrelated steroid receptors in different cellular systems.
Collapse
Affiliation(s)
- Eleanor F Need
- Molecular Ageing Laboratory, The Freemasons Foundation Centre for Men’s Health, Basil Hetzel Institute for Translational Research, Australia
| | | | | | | | | | | |
Collapse
|
14
|
Peters AA, Ingman WV, Tilley WD, Butler LM. Differential effects of exogenous androgen and an androgen receptor antagonist in the peri- and postpubertal murine mammary gland. Endocrinology 2011; 152:3728-37. [PMID: 21846805 DOI: 10.1210/en.2011-1133] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is emerging evidence that androgens inhibit proliferation of normal and malignant breast epithelial cells, but the actions of androgens in normal mammary gland morphogenesis are not well understood. In this study, we investigated whether development of the murine mammary gland could be altered by stimulating or suppressing androgen receptor (AR) signaling in vivo. Intact virgin female mice aged 5 wk (midpuberty) or 12 wk (postpuberty) were implanted with slow-release pellets containing either placebo, 5α-dihydrotestosterone (1.5 mg) or the AR antagonist flutamide (60 mg). Treatment with 5α-dihydrotestosterone from midpuberty to 12 wk of age-retarded ductal extension by 40% (P = 0.007), but treatment from 12-21 wk had no significant effect on gland morphology. In contrast, inhibition of AR signaling with flutamide from midpuberty had no effect on the mammary gland, but flutamide treatment from 12-21 wk increased ductal branching (P = 0.004) and proliferation (P = 0.03) of breast epithelial cells. The increased proliferation in flutamide-treated mice was not correlated with serum estradiol levels or estrogen receptor-α (ERα) expression. In control mice, the frequency and intensity of AR immunostaining in mammary epithelial cells was significantly increased in the 12- to 21-wk treatment group compared with the 5- to 12-wk group (P < 0.001). In contrast, no change in ERα occurred, resulting in a marked increase in the AR to ERα ratio from 0.56 (±0.12) to 1.47 (±0.10). Our findings indicate that androgen signaling influences development and structure of the adult mammary gland and that homeostasis between estrogen and androgen signaling in mature glands is critical to constrain the proliferative effects of estradiol.
Collapse
Affiliation(s)
- A A Peters
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, Hanson Institute, P.O. Box 14 Rundle Mall, Adelaide, South Australia 5000, Australia
| | | | | | | |
Collapse
|
15
|
Kirkpatrick ME, Clark AS. Androgen inhibition of sexual receptivity is modulated by estrogen. Physiol Behav 2010; 102:361-6. [PMID: 21130793 DOI: 10.1016/j.physbeh.2010.11.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 11/03/2010] [Accepted: 11/26/2010] [Indexed: 11/16/2022]
Abstract
Sexual receptivity induced in ovariectomized rats by the long-term administration of estradiol benzoate (EB) can be inhibited by concurrent administration of androgens. Experiment 1 examined the role of time course and dose of androgens in the inhibition of estrogen-induced sexual receptivity. Ovariectomized rats were treated with EB (2.0 microg per rat per day) for 6 days and tested for sexual receptivity (Test Day I). EB treatment continued for 15 days concomitant with daily administration of one of three doses of dihydrotestosterone propionate (DHTP; 7.5, 0.75, 0.075 mg/kg) or 3α-androstanediol (3α-Adiol; 3.75, 1.0, 0.375 mg/kg). Four tests for sexual receptivity were conducted on days 3, 6, 14, and 15 of the androgen/vehicle treatment period (Test Days II-V). On Day 15 (Test Day V), the rats received progesterone (1.0 mg per rat) 4 h before testing. Using the same experimental design, Experiment 2 examined the effect of increasing the dose of estrogen on the androgenic inhibition of sexual receptivity. Ovariectomized rats were treated with one of two doses of EB (2.0 or 10.0 microg per rat per day) concomitant with daily administration of DHTP (7.5 mg/kg) or 3α-Adiol (3.75 mg/kg). In Experiment 1, the highest doses of both DHTP and 3α-Adiol significantly inhibited estrogen-induced sexual receptivity. Data from Experiment 2 indicate that the inhibitory effects of DHTP but not 3α-Adiol can be moderated by an increased dose of EB.
Collapse
Affiliation(s)
- Meg E Kirkpatrick
- Department of Psychology, Wheaton College, Box 18, 26 E. Main Street, Norton, MA 02766, USA.
| | | |
Collapse
|
16
|
Effect of toremifene and ospemifene, compared to acolbifene, on estrogen-sensitive parameters in rat and human uterine tissues. Horm Mol Biol Clin Investig 2010; 1:139-46. [PMID: 25961189 DOI: 10.1515/hmbci.2010.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 10/22/2009] [Indexed: 11/15/2022]
Abstract
BACKGROUND Although the first generation selective estrogen receptor modulator (SERM) tamoxifen (TAM) is well known for its uterotrophic activity, this study compares the stimulatory effect of the TAM derivatives toremifene (TORE) and ospemifene (OSPE) on estrogen-sensitive parameters in rat and human uterine tissues. MATERIAL AND METHODS Ovariectomized female rats were treated daily orally for 10 days with 0.75 mg/rat of TORE, OSPE or acolbifene (ACOL, a pure estrogen antagonist in the uterus and mammary gland), which was used for comparison. Human endometrial carcinoma Ishikawa cells were incubated for 5 days with increasing doses of compounds, in the absence or presence of 1 nM estradiol (E2). RESULTS TORE and OSPE revealed 52% and 56% increases, respectively, in uterine weight, whereas ACOL had no effect. Similar effects were observed on vaginal weight. Endometrial epithelial height increased from 15.82±0.20 to 48.94± 2.12 and 42.14±1.95 μm with TORE and OSPE, respectively, whereas ACOL had no effect. Alkaline phosphatase activity, an estrogen-sensitive parameter in Ishikawa cells, was increased by 144% and 135% with OH-TORE and OH-OSPE, respectively. Owing to their intrinsic estrogenic activity, at maximal concentrations, OH-TORE and OH-OSPE blocked the stimulatory effect of E2 by only 89% compared to 100% with ACOL. CONCLUSIONS The present in vitro and in vivo data show similar stimulatory effects of 4-hydroxytoremifene (OH-TORE) and OH-OSPE on estrogen-sensitive parameters. ACOL, a third generation SERM, has no effect on any of these parameters. Such data add to the potential uterine safety limitations of triphenylethylene-derived SERMs for long-term use in humans.
Collapse
|
17
|
Relative androgen excess during the menopausal transition predicts incident metabolic syndrome in midlife women: study of Women's Health Across the Nation. Menopause 2009; 16:257-64. [PMID: 18971793 DOI: 10.1097/gme.0b013e318185e249] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE During the menopausal transition, total testosterone (T) remains unchanged, whereas estrogen decreases markedly, creating a state of relative androgen excess. We hypothesized that change in the T-to-estradiol (T/E2) ratio during the menopausal transition would be associated with incident metabolic syndrome. METHODS The association between incident metabolic syndrome and total E2, total T, sex hormone-binding globulin, the free androgen index, baseline total T/E2 ratio, and the change of this ratio over time was evaluated in a multiethnic cohort of 1,862 premenopausal and perimenopausal women without diabetes enrolled in the Study of Women's Health Across the Nation. RESULTS New cases (n = 257) of metabolic syndrome were identified in the cohort during 6,296 woman-years of follow-up. The age-adjusted total T/E2 ratio increased by 10.1% per year during the 5 years of follow-up. Neither baseline nor change in E2 was associated with incident metabolic syndrome. Low sex hormone-biding globulin, free androgen index, and high total T at baseline all increased the risk of metabolic syndrome, but their change over time did not. Both baseline total T/E2 ratio (1.41; 95% CI = 1.17-1.69; P < 0.001) and its rate of change (1.24; 95% CI = 1.01-1.52; P < 0.04) were associated with increased incident metabolic syndrome independent of ethnicity. CONCLUSIONS The interaction between T and E2 during the menopausal transition, rather than the individual change of each over time, is a factor in the determination of risk of developing metabolic syndrome during the menopausal transition. This relationship was independent of ethnicity and other factors associated with prevalent metabolic syndrome before the onset of the menopausal transition.
Collapse
|
18
|
Salehi F, Turner MC, Phillips KP, Wigle DT, Krewski D, Aronson KJ. Review of the etiology of breast cancer with special attention to organochlorines as potential endocrine disruptors. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2008; 11:276-300. [PMID: 18368557 DOI: 10.1080/10937400701875923] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Breast cancer is the most frequently diagnosed cancer among Canadian women, accounting for about 30% of all new cancer cases each year. Although the incidence of breast cancer has increased over the past 50 years, the cause of this rise is unknown. Risk factors for breast cancer may be classified into four broad categories: (1) genetic/familial, (2) reproductive/hormonal, (3) lifestyle, and (4) environmental. Established risk factors for breast cancer include older age, later age at first full-term pregnancy, no full-term pregnancies, postmenopausal obesity, and genetic factors. However, these known risk factors cannot account for the majority of cases. In the early 1990s, it was suggested that exposure to some environmental chemicals such as organochlorine compounds may play a causal role in the etiology of breast cancer through estrogen-related pathways. The relationship between organochlorines and breast cancer risk has been studied extensively in the past decade and more, and at this point there is no clear evidence to support a causal role of most organochlorine pesticides in the etiology of human breast cancer, but more evidence is needed to assess risk associated with polychlorinated biphenyls (PCBs). Future studies need to consider the combined effects of exposures, concentrate on vulnerable groups such as those with higher levels of exposure, only consider exposures occurring during the most etiologically relevant time periods, and more thoroughly consider gene-environment interactions.
Collapse
Affiliation(s)
- Fariba Salehi
- McLaughlin Centre for Population Health Risk Assessment, Institute of Population Health, University of Ottawa, Ottawa, Canada
| | | | | | | | | | | |
Collapse
|
19
|
Aubé M, Larochelle C, Ayotte P. 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (p,p'-DDE) disrupts the estrogen-androgen balance regulating the growth of hormone-dependent breast cancer cells. Breast Cancer Res 2008; 10:R16. [PMID: 18275596 PMCID: PMC2374972 DOI: 10.1186/bcr1862] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 12/10/2007] [Accepted: 02/14/2008] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Estrogen and androgen signalling pathways exert opposing influences on the proliferation of mammary epithelial and hormone-dependent breast cancer cells. We previously reported that plasma concentrations of 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (p,p'-DDE), the main metabolite of the insecticide DDT (1,1,1-trichloro-2,2-bis [p-chlorophenyl]ethane) and a potent androgen antagonist, were associated with tumor aggressiveness in women diagnosed with breast cancer. We sought to examine the biological plausibility of this association by testing the effect of p,p'-DDE on the proliferation of CAMA-1 cells, a human breast cancer cell line that expresses the estrogen receptor alpha (ERalpha) and the androgen receptor (AR), in the presence of physiological concentrations of estrogens and androgens in the cell culture medium. METHODS The proliferation of CAMA-1 cells was determined in 96-well plates following a 9-day treatment with p,p'-DDE alone (0.1 to 10 muM) or in combination with 17beta-estradiol (E2) (100 pM) and dihydrotestosterone (DHT) (100, 500, or 1,000 pM). We also assessed p,p'-DDE-induced modifications in cell cycle entry and the expression of the sex-steroid-dependent genes ESR1, AR, CCND1, and TFF1 (pS2) (mRNA and/or protein). RESULTS We found that treatment with p,p'-DDE induced a dose-response increase in the proliferation of CAMA-1 cells when cultivated in the presence of physiological concentrations of estrogens and androgens, but not in the absence of sex steroids in the cell culture medium. A similar effect of p,p'-DDE was noted on the proliferation of MCF7-AR1 cells, an estrogen-responsive cell line that was genetically engineered to overexpress the AR. DHT added together with E2 to the cell culture medium decreased the recruitment of CAMA-1 cells in the S phase and the expression of ESR1 and CCND1 by comparison with cells treated with E2 alone. These androgen-mediated effects were blocked with similar efficacy by p,p'-DDE and the potent antiandrogen hydroxyflutamide. CONCLUSION Our results suggest that p,p'-DDE could increase breast cancer progression by opposing the androgen signalling pathway that inhibits growth in hormone-responsive breast cancer cells. The potential role of environmental antiandrogens in breast carcinogenesis deserves further investigation.
Collapse
Affiliation(s)
- Michel Aubé
- Unité de Recherche en Santé Publique, Centre de Recherche du Centre Hospitalier Universitaire de Québec-CHUL, 2875 boulevard Laurier, Québec, QC G1V 2M2, Canada
| | | | | |
Collapse
|
20
|
Birrell SN, Butler LM, Harris JM, Buchanan G, Tilley WD. Disruption of androgen receptor signaling by synthetic progestins may increase risk of developing breast cancer. FASEB J 2007; 21:2285-93. [PMID: 17413000 DOI: 10.1096/fj.06-7518com] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There is now considerable evidence that using a combination of synthetic progestins and estrogens in hormone replacement therapy (HRT) increases the risk of breast cancer compared with estrogen alone. Furthermore, the World Health Organization has recently cited combination contraceptives, which contain synthetic progestins, as potentially carcinogenic to humans, particularly for increased breast cancer risk. Given the above observations and the current trend toward progestin-only contraception, it is important that we have a comprehensive understanding of how progestins act in the millions of women worldwide who regularly take these medications. While synthetic progestins, such as medroxyprogesterone acetate (MPA), which are currently used in both HRT and oral contraceptives were designed to act exclusively through the progesterone receptor, it is clear from both clinical and experimental settings that their effects may be mediated, in part, by binding to the androgen receptor (AR). Disruption of androgen action by synthetic progestins may have serious deleterious side effects in the breast, where the balance between estrogen signaling and androgen signaling plays a critical role in breast homeostasis. Here, we review the role of androgen signaling in the normal breast and in breast cancer and present new data demonstrating that androgen receptor function can be perturbed by low doses of MPA, similar to doses achieved in serum of women taking HRT. We propose that the observed excess of breast malignancies associated with combined HRT may be explained, in part, by synthetic progestins such as MPA acting as endocrine disruptors to negate the protective effects of androgen signaling in the breast. Understanding the role of androgen signaling in the breast and how this is modulated by synthetic progestins is necessary to determine how combined HRT alters breast cancer risk, and to inform the development of optimal preventive and treatment strategies for this disease.
Collapse
Affiliation(s)
- Stephen N Birrell
- Dame Roma Mitchell Cancer Research Laboratories, The University of Adelaide, Hanson Institute, PO Box 14, Rundle Mall, South Australia, 5000, Australia
| | | | | | | | | |
Collapse
|
21
|
Bhattacharyya RS, Krishnan AV, Swami S, Feldman D. Fulvestrant (ICI 182,780) down-regulates androgen receptor expression and diminishes androgenic responses in LNCaP human prostate cancer cells. Mol Cancer Ther 2006; 5:1539-49. [PMID: 16818513 DOI: 10.1158/1535-7163.mct-06-0065] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The androgen receptor (AR) plays a key role in the development and progression of prostate cancer. Targeting the AR for down-regulation would be a useful strategy for treating prostate cancer, especially hormone-refractory or androgen-independent prostate cancer. In the present study, we showed that the antiestrogen fulvestrant [ICI 182,780 (ICI)] effectively suppressed AR expression in several human prostate cancer cells, including androgen-independent cells. In LNCaP cells, ICI (10 micromol/L) treatment decreased AR mRNA expression by 43% after 24 hours and AR protein expression by approximately 50% after 48 hours. We further examined the mechanism of AR down-regulation by ICI in LNCaP cells. ICI did not bind to the T877A-mutant AR present in the LNCaP cells nor did it promote proteasomal degradation of the AR. ICI did not affect AR mRNA or protein half-life. However, ICI decreased the activity of an AR promoter-luciferase reporter plasmid transfected into LNCaP cells, suggesting a direct repression of AR gene transcription. As a result of AR down-regulation by ICI, androgen induction of prostate-specific antigen mRNA and protein expression were substantially attenuated. Importantly, LNCaP cell proliferation was significantly inhibited by ICI treatment. Following 6 days of ICI treatment, a 70% growth inhibition was seen in androgen-stimulated LNCaP cells. These data show that the antiestrogen ICI is a potent AR down-regulator that causes significant inhibition of prostate cancer cell growth. Our study suggests that AR down-regulation by ICI would be an effective strategy for the treatment of all prostate cancer, especially AR-dependent androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Rumi S Bhattacharyya
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5103, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Mainly through the transformation of dehydroepiandrosterone (DHEA) into androgens in peripheral tissues by intracrine mechanisms, women synthesize at least two-thirds of the androgens found in men. Such data strongly suggest that androgens exert very important but so far underestimated physiological functions in women, including in the breast. In fact, the mammary gland possesses all the enzymatic machinery required to transform DHEA into both androgens and estrogens, although androgens are the predominant steroids synthesized from DHEA in the mammary gland. Early clinical studies have shown beneficial effects of androgens on breast cancer which are comparable to those observed with other hormonal therapies. In fact, a long series of preclinical and clinical data clearly indicate that proliferation of both the normal mammary gland and breast cancer results from the balance between the stimulatory effect of estrogens and the inhibitory effect of androgens. Moreover, the data showing the additive inhibitory effects of antiestrogens and androgens suggest that taking advantage of the inhibitory effect of androgens on breast cancer proliferation could well improve the efficacy of the currently used estrogen deprivation therapies for the treatment and prevention of breast cancer, the best and most physiological candidate being DHEA that limits the androgenic exposure to the tissues which possess the required enzymatic intracrine machinery.
Collapse
Affiliation(s)
- Fernand Labrie
- Laboratory of Molecular Endocrinology and Oncology, Laval University Hospital Research Center (CRCHUL) and Laval University, Quebec City, Quebec, Canada.
| |
Collapse
|
23
|
Durrer S, Maerkel K, Schlumpf M, Lichtensteiger W. Estrogen target gene regulation and coactivator expression in rat uterus after developmental exposure to the ultraviolet filter 4-methylbenzylidene camphor. Endocrinology 2005; 146:2130-9. [PMID: 15705771 DOI: 10.1210/en.2004-1272] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Because the estrogen receptor (ER) ligand type influences transactivation, it is important to obtain information on molecular actions of nonclassical ER agonists. UV filters from cosmetics represent new classes of endocrine active chemicals, including the preferential ER beta ligands 4-methylbenzylidene camphor (4-MBC) and 3-benzylidene camphor. We studied estrogen target gene expression in uterus of Long Evans rats after developmental exposure to 4-MBC (0.7, 7, 24, and 47 mg/kg x d) administered in feed to the parent generation before mating, during pregnancy and lactation, and to the offspring until adulthood. 4-MBC altered steady-state levels of mRNAs encoding for ER alpha, ER beta, progesterone receptor (PR), IGF-I, androgen receptor, determined by real-time RT-PCR in uterus of 12-wk-old offspring. Western-blot analyses of the same tissue homogenates indicated changes in ER alpha and PR but not ER beta proteins. To assess sensitivity to estradiol (E2), offspring were ovariectomized on d 70, injected with E2 (10 or 50 microg/kg sc) on d 84, and killed 6 h later. Acute up-regulation of PR and IGF-I and down-regulation of ER alpha and androgen receptor by E2 were dose-dependently reduced in 4-MBC-exposed rats. The reduced response to E2 was accompanied by reduced coactivator SRC-1 mRNA and protein levels. Our data indicate that developmental exposure to 4-MBC affects the regulation of estrogen target genes and the expression of nuclear receptor coregulators in uterus at mRNA and protein levels.
Collapse
Affiliation(s)
- Stefan Durrer
- Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
| | | | | | | |
Collapse
|
24
|
Saaresranta T, Aittokallio T, Utriainen K, Polo O. Medroxyprogesterone improves nocturnal breathing in postmenopausal women with chronic obstructive pulmonary disease. Respir Res 2005; 6:28. [PMID: 15807890 PMCID: PMC1079947 DOI: 10.1186/1465-9921-6-28] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2005] [Accepted: 04/04/2005] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Progestins as respiratory stimulants in chronic obstructive pulmonary disease (COPD) have been investigated in males and during wakefulness. However, sleep and gender may influence therapeutic responses. We investigated the effects of a 2-week medroxyprogesterone acetate (MPA) therapy on sleep and nocturnal breathing in postmenopausal women. METHODS A single-blind placebo-controlled trial was performed in 15 postmenopausal women with moderate to severe COPD. A 12-week trial included 2-week treatment periods with placebo and MPA (60 mg/d/14 days). All patients underwent a polysomnography with monitoring of SaO2 and transcutaneous PCO2 (tcCO2) at baseline, with placebo, with medroxyprogesterone acetate (MPA 60 mg/d/14 days), and three and six weeks after cessation of MPA. RESULTS Thirteen patients completed the trial. At baseline, the average +/- SD of SaO2 mean was 90.6 +/- 3.2 % and the median of SaO2 nadir 84.8 % (interquartile range, IQR 6.1). MPA improved them by 1.7 +/- 1.6 %-units (95 % confidence interval (CI) 0.56, 2.8) and by 3.9 %-units (IQR 4.9; 95% CI 0.24, 10.2), respectively. The average of tcCO2 median was 6.0 +/- 0.9 kPa and decreased with MPA by 0.9 +/- 0.5 kPa (95% CI -1.3, -0.54). MPA improved SaO2 nadir and tcCO2 median also during REM sleep. Three weeks after cessation of MPA, the SaO2 mean remained 1.4 +/- 1.8 %-units higher than at baseline, the difference being not significant (95% CI -0.03, 2.8). SaO2 nadir was 2.7 %-units (IQR 4.9; 95% CI 0.06, 18.7) higher than at baseline. Increases in SaO2 mean and SaO2 nadir during sleep with MPA were inversely associated with baseline SaO2 mean (r = -0.70, p = 0.032) and baseline SaO2 nadir (r = -0.77, p = 0.008), respectively. Treatment response in SaO2 mean, SaO2 nadir and tcCO2 levels did not associate with pack-years smoked, age, BMI, spirometric results or sleep variables. CONCLUSION MPA-induced respiratory improvement in postmenopausal women seems to be consistent and prolonged. The improvement was greater in patients with lower baseline SaO2 values. Long-term studies in females are warranted.
Collapse
Affiliation(s)
- Tarja Saaresranta
- Sleep Research Unit at the Department of Physiology, University of Turku, Sleep Research Unit, Dentalia, Lemminkäisenkatu 2, 20520 Turku, Finland
- Department of Pulmonary Diseases, Turku University Central Hospital, 20520 Turku, Finland
| | - Tero Aittokallio
- Department of Mathematics, University of Turku, 20014 Turku, Finland
| | - Karri Utriainen
- Sleep Research Unit at the Department of Physiology, University of Turku, Sleep Research Unit, Dentalia, Lemminkäisenkatu 2, 20520 Turku, Finland
| | - Olli Polo
- Sleep Research Unit at the Department of Physiology, University of Turku, Sleep Research Unit, Dentalia, Lemminkäisenkatu 2, 20520 Turku, Finland
- Department of Pulmonary Diseases, Turku University Central Hospital, 20520 Turku, Finland
- Department of Pulmonary Diseases, Tampere University Central Hospital, P.O.Box 2000, 33521 Tampere, Finland
| |
Collapse
|
25
|
Leão LMCSM, Duarte MPC, Farias MLF. Insuficiência androgênica na mulher e potenciais riscos da reposição terapêutica. ACTA ACUST UNITED AC 2005; 49:205-16. [PMID: 16184248 DOI: 10.1590/s0004-27302005000200006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Na mulher, os androgênios decrescem lenta e progressivamente a partir da quarta década e por toda a vida. O declínio dos androgênios pode gerar um estado de deficiência que se manifesta insidiosamente por diminuição da função sexual, bem estar e energia, alterações na composição corporal e perda de massa óssea. Se há história de ooforectomia bilateral, pan-hipopituitarismo, supressão da androgênese adrenal e/ou os níveis séricos de testosterona biodisponível se encontram reduzidos, é provável que estes sinais e sintomas sejam aliviados pela administração criteriosa de androgênios, cuja prática tem se difundido. Nas doses atualmente preconizadas, parece que os benefícios sobre massa óssea, sexualidade e qualidade de vida são alcançados sem importantes efeitos colaterais de virilização. Entretanto, trabalhos bem controlados são necessários para validar a hipótese de que a administração terapêutica de androgênios em mulheres não tem, a longo prazo, repercussões significativas na incidência sobre câncer de mama ou conseqüências metabólicas indesejáveis.
Collapse
|
26
|
Lanzino M, De Amicis F, McPhaul MJ, Marsico S, Panno ML, Andò S. Endogenous coactivator ARA70 interacts with estrogen receptor alpha (ERalpha) and modulates the functional ERalpha/androgen receptor interplay in MCF-7 cells. J Biol Chem 2005; 280:20421-30. [PMID: 15772083 DOI: 10.1074/jbc.m413576200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Overexpression of androgen receptor (AR) decreases estrogen receptor alpha (ERalpha) transactivation, which plays a basic role in hormone-dependent breast cancer. This transcriptional interference can be due to shared coactivators. Here we demonstrated that in MCF-7 cells ARA70, an AR-specific coactivator, interacted with endogenous ERalpha, increasing its transcriptional activity, and it was recruited to the pS2 gene promoter. Moreover, a dominant negative ARA70 down-regulated ERalpha transcriptional activity as well as pS2 mRNA. ARA70 overexpression reversed the AR down-regulatory effect on ERalpha signaling. However, in the presence of a progressive increase of transfected AR, ARA70 switched into enhancing the inhibitory effect of AR on ERalpha signaling. These opposite effects of ARA70 were further evidenced by coimmunoprecipitation assay in MCF-7wt, MCF-7-overexpressing AR, and HeLa cells, exogenously expressing an excess of ERalpha with respect to AR or an excess of AR with respect to ERalpha. Thus, ARA70 is a coactivator for ERalpha and may represent a functional link between ERalpha/AR modulating their cross-talk in models of estrogen signaling in MCF-7 and HeLa cells.
Collapse
Affiliation(s)
- Marilena Lanzino
- Department of Pharmacobiology, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | | | | | | | | | | |
Collapse
|
27
|
Affiliation(s)
- Ralph F Fregosi
- Department of Physiology, College of Medicine, University of Arizona, Tucson 85721-0093, USA.
| |
Collapse
|
28
|
Stroheker T, Picard K, Lhuguenot JC, Canivenc-Lavier MC, Chagnon MC. Steroid activities comparison of natural and food wrap compounds in human breast cancer cell lines. Food Chem Toxicol 2004; 42:887-97. [PMID: 15110097 DOI: 10.1016/j.fct.2004.01.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2003] [Revised: 01/16/2004] [Accepted: 01/23/2004] [Indexed: 11/27/2022]
Abstract
In this study, we tested and compared the endocrine disruption activities of compounds in materials used to package foods (bisphenol A, bisphenol F, and bisphenol A diglycidylether BADGE) with natural molecules (genistein, apigenin, kaempferol, and tangeretin) in the human breast cancer cell lines MCF-7 (ER(+)) and MDA-MB453 (AR(+); GR(+)). Octylphenol was also chosen as a xenoestrogen reference. Two compounds had no estrogenic activity: BADGE and tangeretin. Genistein was the most active compound in the E-Screen assay with MCF-7, followed by octylphenol, bisphenol F, bisphenol A and apigenin, with kaempferol the least potent. All estrogenic compounds competed with 17beta-estradiol for binding to the MCF-7 ER and their estrogenic effects were abolished in the presence of tamoxifen, an ER antagonist. In MDA-MB453 cells transfected with pMMTVneo-Luc, all compounds had anti-androgenic activities, with octylphenol the most potent. Kaempferol, genistein, and apigenin were more potent anti-androgens than bisphenols A or F. The natural compounds had a biphasic effect on luciferase activity. At high concentrations, genistein (10(-5)M) and apigenin (10(-6)M) acted as GR agonists in transfected MDA-MB453 cells. Furthermore, apigenin, at a concentration of 10(-5)M, may act as a partial androgen receptor (AR) agonist, as nilutamide, an AR antagonist, inhibited its activity by 26%.
Collapse
Affiliation(s)
- T Stroheker
- UMR 1234 Toxicologie Alimentaire, University of Burgundy/INRA, 1 Esplanade Erasme, 21000 Dijon, France
| | | | | | | | | |
Collapse
|
29
|
Saaresranta T, Polo O. Long-term progestin therapy for female chronic respiratory insufficiency? Respir Med 2004; 98:194-5. [PMID: 14971885 DOI: 10.1016/j.rmed.2003.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
End-stage chronic obstructive pulmonary disease often leads to hypercapnic respiratory failure. Oxygen supplementation therapy may further aggravate hypercapnia and not all patients are compliant with non-invasive ventilation. This case documents successful control of chronic respiratory failure with medroxyprogesterone in a postmenopausal woman during 1-year follow-up.
Collapse
Affiliation(s)
- Tarja Saaresranta
- Department of Pulmonary Diseases, Turku University Central Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland.
| | | |
Collapse
|
30
|
Abstract
The role of androgens in women's health has been generally neglected. Currently available assays are lacking in sensitivity and reliability at the lower ranges. Circulating androgens as prohormones for other steroids (e.g., androgens) have an ubiquitous role on diverse physiological and behavioral systems. Clinical assessment of both androgen production and androgen availability can be achieved by measurement of two or three essential values. These include either total T and sex-hormone binding globulin (SHBG), free T and SHBG, or free T and total T. The free testosterone index (total T/SHBG) correlates well with free or bioavailable T and can be used as a substitute. DHEA-S is the most useful measure of adrenal androgen production in women. Androgen insufficiency in women is not a specific consequence of natural menopause, but may occur secondarily to the age-related decline in both adrenal and ovarian androgen production. Since estrogen effects are also strongly linked to mood, psychological well-being, and sexual function in women, the diagnosis of androgen insufficiency should only be made in women who are adequately estrogenized. Before initiating a trial of androgen replacement therapy, a comprehensive clinical assessment should be performed in all cases. Approved androgen replacement therapy is not yet available in most countries for treatment of female sexual dysfunction. This would include T supplements or DHEA. Several new progestins have been synthesized in the last decade. Dienogest is a hybrid progestin that is derived from both the pregnane and the estrane groups with a 17alpha-cyanomethyl radical; drospirenone is derived from spirolactone. Somehow, the molecules available have demonstrated antiandrogenic properties. Cyproterone acetate (CPA) is the most potent antiandrogenic progestin, followed by dienogest, drosperinone, and chlormadinone acetate. Nomegestrol acetate and medrogestone also exert some antiandrogenic properties and are similar to chlormadinone acetate in antiandrogenic potency. While androgens act positively on libido in women, antiandrogenic properties in doses used in HRT do not appear to have a negative effect. Progestins used in HRT have varying pharmacological properties that are associated with different adverse effects. The new progestins with antiandrogenic properties avoid many of the androgenic side effects related to testosterone-derived progestins. They also have the potential of not counteracting beneficial estrogen effects, for example, on the cardiovascular system or mental tone.
Collapse
Affiliation(s)
- H P G Schneider
- Department of Obstetrics Gynecology, University of Muenster, Von-Esmarch-Strasse 56, ZMBE, 48149 Muenster, Germany.
| |
Collapse
|
31
|
Stroheker T, Cabaton N, Berges R, Lamothe V, Lhuguenot JC, Chagnon MC. Influence of dietary soy isoflavones on the accessory sex organs of the Wistar rat. Food Chem Toxicol 2003; 41:1175-83. [PMID: 12842186 DOI: 10.1016/s0278-6915(03)00108-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We evaluated the effects of three rodent diets differing in soybean meal content on the response of the seminal vesicles, prostate and bulbocavernosus/levator ani (BC/LA) muscle to androgens and anti-androgenic compounds in the Hershberger assay. The diets tested were (1) L5, a semi-synthetic phytoestrogen-free diet, (2) DO4, 8.5% (w/w) vegetable protein and (3) DO3, 22.5% (w/w) vegetable protein. We determined the effects of dietary soy isoflavones after ten days of exposure and in animals fed L5 and DO3 diets throughout their lifetime (including the period of treatment with androgenic or anti-androgenic compounds). After ten days of exposure, we observed no effect of diet on the accessory sex organs of male Wistar rats. In contrast, diet affected the androgenic response to testosterone propionate in seminal vesicles and prostate. Seminal vesicles were the most sensitive organs. Vinclozolin caused a dose-dependent decrease in the relative weights of seminal vesicles, prostate and BC/LA regardless of diet. As vegetable proteins may contain high proportions of genistein and daidzein, two well-known oestrogenic endocrine disrupters that may alter the results of reproductive studies, we recommend the use of a standardised open-formula diet without soy isoflavones, such as L5, if the Hershberger assay is to be performed.
Collapse
Affiliation(s)
- Thomas Stroheker
- UMR 0938 de Toxicologie Alimentaire, Ensbana, 1 esplanade Erasme, 21000, Dijon, France
| | | | | | | | | | | |
Collapse
|
32
|
Labrie F, Luu-The V, Labrie C, Bélanger A, Simard J, Lin SX, Pelletier G. Endocrine and intracrine sources of androgens in women: inhibition of breast cancer and other roles of androgens and their precursor dehydroepiandrosterone. Endocr Rev 2003; 24:152-82. [PMID: 12700178 DOI: 10.1210/er.2001-0031] [Citation(s) in RCA: 377] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Serum androgens as well as their precursors and metabolites decrease from the age of 30-40 yr in women, thus suggesting that a more physiological hormone replacement therapy at menopause should contain an androgenic compound. It is important to consider, however, that most of the androgens in women, especially after menopause, are synthesized in peripheral intracrine tissues from the inactive precursors dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEA-S) of adrenal origin. Much progress in this new area of endocrine physiology called intracrinology has followed the cloning and characterization of most of the enzymes responsible for the transformation of DHEA and DHEA-S into androgens and estrogens in peripheral target tissues, where the locally produced sex steroids are exerting their action in the same cells in which their synthesis takes place without significant diffusion into the circulation, thus seriously limiting the interpretation of serum levels of active sex steroids. The sex steroids made in peripheral tissues are then inactivated locally into more water-soluble compounds that diffuse into the general circulation where they can be measured. In a series of animal models, androgens and DHEA have been found to inhibit breast cancer development and growth and to stimulate bone formation. In clinical studies, DHEA has been found to increase bone mineral density and to stimulate vaginal maturation without affecting the endometrium, while improving well-being and libido with no significant side effects. The advantage of DHEA over other androgenic compounds is that DHEA, at physiological doses, is converted into androgens and/or estrogens only in the specific intracrine target tissues that possess the appropriate physiological enzymatic machinery, thus limiting the action of the sex steroids to those tissues possessing the tissue-specific profile of expression of the genes responsible for their formation, while leaving the other tissues unaffected and thus minimizing the potential side effects observed with androgens or estrogens administered systemically.
Collapse
Affiliation(s)
- Fernand Labrie
- Molecular Endocrinology and Oncology Research Center, Laval University Medical Center (Centre Hospitalier de l'Université Laval) and Laval University, Québec City, Québec G1V 4G2, Canada.
| | | | | | | | | | | | | |
Collapse
|
33
|
Nomura M, Korach KS, Pfaff DW, Ogawa S. Estrogen receptor beta (ERbeta) protein levels in neurons depend on estrogen receptor alpha (ERalpha) gene expression and on its ligand in a brain region-specific manner. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 110:7-14. [PMID: 12573528 DOI: 10.1016/s0169-328x(02)00544-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Estrogen receptors (ERs) alpha and beta are very similar estrogen-binding proteins, perhaps gene duplication products, which act as ligand-dependent transcription factors. While the estrogenic regulation of ERalpha has been well documented, little is known about how estrogen regulates ERbeta and whether ERalpha plays a role in the expression and estrogenic regulation of ERbeta. In the present study, we examined the effects of gonadectomy and estrogen replacement on ERbeta immunoreactivity (ir) in wild-type (WT) and ERalpha knockout (alphaERKO) adult male mice in six brain regions, the medial preoptic area (MPOA), the bed nucleus of the stria terminalis (BNST), the paraventricular nucleus (PVN), the ventromedial nucleus of hypothalamus (VMH), the medial amygdala nucleus (MeAMY) and the dorsal raphe nucleus (DRN). Mice were divided into four different treatment groups: gonadally intact, gonadectomized (GDX), GDX+short-term treatment with estrogen (s.c. injection of estradiol benzoate (EB), 5 microg, at 48 h before perfusion) or GDX+long-term treatment with estrogen (implant of an EB pellet, 2.5 microg/day, for 10 days). In intact alphaERKO mice, the number of ERbeta expressing cells was significantly decreased in the MPOA and increased in the BNST, compared to WT mice. Both in the MPOA and BNST, steroid hormone regulation of ERbeta protein (an increase by GDX and a decline to intact levels by EB) was found only in WT, not in alphaERKO mice. In the VMH, GDX significantly increased the number of ERbeta ir expressing cells in both genotypes. EB treatment tended to decrease the number of ERbeta ir cells in WT mice, whereas EB treatment tended to increase ERbeta ir cell counts in alphaERKO mice. No effects of GDX or EB treatment were found in the DRN and MeAMY regardless of genotype. These results suggest that gonadal steroid hormones may regulate ERbeta protein in male mice and ERalpha may be involved in the expression and regulation of ERbeta in a region-specific manner.
Collapse
Affiliation(s)
- Masayoshi Nomura
- Laboratory of Neurobiology and Behavior, The Rockefeller University, Box 275, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
34
|
Custodia-Lora N, Novillo A, Callard IP. Effect of gonadal steroids on progesterone receptor, estrogen receptor, and vitellogenin expression in male turtles (Chrysemys picta). ACTA ACUST UNITED AC 2003; 301:15-25. [PMID: 14695685 DOI: 10.1002/jez.a.20004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hepatic vitellogenin (vtg) is a yolk precursor protein sequestered in follicular oocytes as nutrient supply for developing embryos in nonmammalian vertebrates. In prior research studies we have demonstrated that both progesterone (P) and testosterone (T) inhibit estrogen (E)-induced vitellogenesis in the male fresh water turtle (Chrysemys picta), and have suggested that these hormones may be involved in multihormonal regulation of vitellogenesis in the female turtle. However, the modes of action of progesterone and testosterone on estrogen-induced vitellogenesis are not known. We have proposed that progesterone inhibits vitellogenesis by modulation of progesterone receptor A (PRA) or B (PRB) isoforms and/or estrogen receptor (ER) gene transcription. In this study, we compare the vitellogenic responses of reproductively inactive male turtles to estradiol 17beta in the presence of exogenous testosterone or progesterone. Northern blot analysis was used to monitor the changes in vtg mRNA, ER mRNA, and PR mRNA expression; Western blotting to determine changes in PR isoform expression and a homologous ELISA for measurement of plasma vtg. Progesterone and testosterone reduced estrogen-induced vtg mRNA expression, but plasma vtg was not significantly reduced by these steroids. PRA and PRB were transcribed even though ER mRNA could not be detected, suggesting constitutive PR expression. However, in the presence of estradiol 17beta, both PR isoforms and mRNA transcripts were increased as a correlate of ER mRNA transcription, suggesting both transcriptional and translational effects; these effects were inhibited by testosterone and progesterone treatments. Since ER mRNA was sharply reduced by both testosterone and progesterone, and estradiol 17 beta increased PR mRNA transcription and translation, it is likely that the action of progesterone in reducing vtg mRNA is indirect via down regulation of ER mRNA, thus ER. This study provides further information on the role of progesterone and testosterone in the regulation of hepatic vitellogenesis, suggesting regulation of vitellogenesis mainly via modulation of hepatic ER mRNA.
Collapse
|
35
|
Abstract
A number of hormones, including hypothalamic neuropeptides acting as neurotransmitters and neuromodulators in the CNS, are involved in the physiologic regulation of breathing and participate in adjustment of breathing in disease. In addition to central effects, some hormones also control breathing at peripheral chemoreceptors or have local effects on the lungs and airways. Estrogen and progesterone seem to protect from sleep-disordered breathing, whereas testosterone may predispose to it. Progesterone and thyroxine have long been known to stimulate respiration. More recently, several hormones such as corticotropin-releasing hormone and leptin have been suggested to act as respiratory stimulants. Somatostatin, dopamine, and neuropeptide Y have a depressing effect on breathing. Animal models and experimental human studies suggest that also many other hormones may be involved in respiratory control.
Collapse
Affiliation(s)
- Tarja Saaresranta
- Department of Pulmonary Diseases, Turku University Central Hospital, Kiinamyllynkatu 4-8, FIN-20520 Turku, Finland.
| | | |
Collapse
|
36
|
Carsol JL, Gingras S, Simard J. Synergistic action of prolactin (PRL) and androgen on PRL-inducible protein gene expression in human breast cancer cells: a unique model for functional cooperation between signal transducer and activator of transcription-5 and androgen receptor. Mol Endocrinol 2002; 16:1696-710. [PMID: 12089361 DOI: 10.1210/mend.16.7.0875] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The signal transducer and activator of transcription 5 (Stat5) has been shown to cooperate with some nuclear receptors. However, an interaction has never been demonstrated with the androgen receptor (AR). Given that the PRL-inducible protein/gross cystic disease fluid-15 (PIP/GCDFP-15) is both a PRL-controlled and an androgen-controlled protein, we used its promoter region to investigate the potential interaction between Stat5 and androgen receptor. Dihydrotestosterone or PRL alone slightly modulated or did not modulate the luciferase activity of all reporter gene constructs. In contrast, a maximal increase was observed using the -1477+42 reporter gene construct after exposure to both dihydrotestosterone and PRL. The requirement of half-site androgen-responsive elements and two consensus Stat5-binding elements, Stat5#1 and Stat5#2, was determined by site-directed mutagenesis. Activated Stat5B binds with a higher affinity to Stat5#2 than to Stat5#1. Stat5ADelta749 and Stat5BDelta754 mutants demonstrated that the Stat5 trans-activation domain is involved in the hormonal cooperation. The cooperation depends on the PRL-induced phosphorylation on Tyr(694) in Stat5A and Tyr(699) in Stat5B, as demonstrated using the Stat5AY694F and Stat5BY699F proteins. The use of AR Q798E, C619Y, and C784Y mutants showed that trans-activation, DNA-binding, and ligand-binding domains of AR are essential. Our study thus suggests a functional cooperation between AR and Stat5.
Collapse
Affiliation(s)
- Jean-Louis Carsol
- Canada Research Chair in Oncogenetics, Oncology and Molecular Endocrinology Research Center, Laval University Medical Center and Laval University, Québec, Canada G1V 4G2
| | | | | |
Collapse
|
37
|
Abstract
OBJECTIVE Evaluation of current clinical, experimental, genetic, and epidemiological data pertaining to the role of androgens in mammary growth and neoplasia. DESIGN Literature review. SETTING National Institutes of Health. SUBJECT(S) Recent, basic, clinical, and epidemiological studies. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Effects of androgens on mammary epithelial proliferation and/or breast cancer incidence. RESULT(S) Experimental data derived from rodents and cell lines provide conflicting results that appear be strain- and cell line-dependent. Epidemiologic studies have significant methodological limitations and provide inconclusive results. The study of molecular defects involving androgenic pathways in breast cancer is in its infancy. Clinical and nonhuman primate studies, however, suggest that androgens inhibit mammary epithelial proliferation and breast growth and that conventional estrogen treatment suppresses endogenous androgens. CONCLUSION(S) Abundant clinical evidence suggests that androgens normally inhibit mammary epithelial proliferation and breast growth. Suppression of androgens by conventional estrogen treatment may thus enhance estrogenic breast stimulation and possibly breast cancer risk. Clinical trials to evaluate the impact of combined estrogen and androgen hormone replacement regimens on mammary gland homeostasis are needed to address this issue.
Collapse
Affiliation(s)
- Constantine Dimitrakakis
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
38
|
Labrie F, Labrie C, Bélanger A, Giguere V, Simard J, Mérand Y, Gauthier S, Luu-The V, Candas B, Martel C, Luo S. Pure selective estrogen receptor modulators, new molecules having absolute cell specificity ranging from pure antiestrogenic to complete estrogen-like activities. ADVANCES IN PROTEIN CHEMISTRY 2001; 56:293-368. [PMID: 11329857 DOI: 10.1016/s0065-3233(01)56009-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Affiliation(s)
- F Labrie
- Oncology and Molecular Endocrinology Research Center, Laval University Medical Center (CHUL), Québec, G1V 4G2, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
There is evidence suggesting that local intracrine formation of sex steroids from inactive precursors, dehydroepiandrosterone (DHEA), its sulfate (DHEA-S) and 4-androstenedione (4-DIONE) plays an important role in the regulation of growth and function of peripheral target tissues. Moreover, human solid tumors are often infiltrated by stromal/immune cells secreting a wide spectra of cytokines. These cytokines might in turn regulate the activity of both immune and neoplastic cells. Our data demonstrate that the potent regulatory effects of interleukin-4 (IL-4) and IL-6 on both estrogenic and androgenic 17beta-HSD/KSR activities in breast cancer cells depend on the cell-specific gene expression of various types of 17beta-HSD/KSR enzymes. However, in both estrogen-receptor (ER)-positive (ZR-75-1, T-47D) and ER-negative (MDA-MB-231, BT-20) human breast cancer cells, exposure to IL-4 and IL-13 caused a rapid and potent induction of 3beta-HSD type 1 gene expression. Such an induction was also observed in normal human mammary and prostate epithelial cells in primary culture as well as in human HaCaT immortalized keratinocytes, ME-180 cervix cancer cells, and HT-29 colon cancer cells. The DNA-binding activity of Stat6, a member of the Signal Transducers and Activators of Transcription gene family, was activated after a 30 min exposure to IL-4 in all the cell types where IL-4 induced 3beta-HSD expression, but not in those that failed to respond to IL-4. Our data therefore suggest that IL-4 and IL-13 may play a role in the biosynthesis of active sex steroids from the inactive adrenal steroid DHEA, not only in breast cells but also in various cell types derived from peripheral target tissues.
Collapse
Affiliation(s)
- J Simard
- Laboratory of Hereditary Cancers, Oncology and Molecular Endocrinology Research Center, Laval University Medical Center (CHUL) and Laval University, 2705 Laurier Boulevard, G1V 4G2, Québec, Canada.
| | | |
Collapse
|
40
|
Panet-Raymond V, Gottlieb B, Beitel LK, Pinsky L, Trifiro MA. Interactions between androgen and estrogen receptors and the effects on their transactivational properties. Mol Cell Endocrinol 2000; 167:139-50. [PMID: 11000528 DOI: 10.1016/s0303-7207(00)00279-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The physiological interplay of androgen and estrogen action in endocrine tissues is well recognized. The biochemical processes responsible for this interplay have yet to be fully defined. We have demonstrated that the androgen receptor (AR) and estrogen receptor-alpha (ERalpha) can interact directly using the yeast and mammalian two-hybrid systems. These interactions occurred between the C-terminal ERalpha ligand-binding domain and either the N-terminal AR transactivational domain or the full-length AR. Estrogen receptor-beta (ERbeta) did not interact with the AR. DNA cotransfection studies employing AR, ERalpha and ERbeta expression vectors and AR- or ER-reporter gene constructs were used to identify and measure potential functional effects of AR-ER interaction. Coexpression of ERalpha with AR decreased AR transactivation by 35%; coexpression of AR with ERalpha decreased ERalpha transactivation by 74%. Coexpression of AR and ERbeta did not significantly modulate AR or ERbeta transactivation. In summary, we have shown that specific domains of AR and ERalpha physically interact and have demonstrated the functional consequences of such interaction. These results may help explain the nature of the physiological interplay between androgens and estrogens.
Collapse
Affiliation(s)
- V Panet-Raymond
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, 3755 Cote-Ste-Catherine Road, Quebec, H3T 1E2, Montreal, Canada
| | | | | | | | | |
Collapse
|
41
|
Prolonged Endocrine Responses to Medroxyprogesterone in Postmenopausal Women With Respiratory Insufficiency. Obstet Gynecol 2000. [DOI: 10.1097/00006250-200008000-00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Diel P, Schulz T, Smolnikar K, Strunck E, Vollmer G, Michna H. Ability of xeno- and phytoestrogens to modulate expression of estrogen-sensitive genes in rat uterus: estrogenicity profiles and uterotropic activity. J Steroid Biochem Mol Biol 2000; 73:1-10. [PMID: 10822019 DOI: 10.1016/s0960-0760(00)00051-0] [Citation(s) in RCA: 169] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The function of the uterus is regulated by female sex steroids and it is, therefore, used as the classical target organ to detect estrogenic action. Uterine response to estrogens involves the activation of a large pattern of estrogen-sensitive genes. This fact offers the opportunity to analyze the estrogenic activity of xeno- and phytoestrogens, and the mechanisms of their molecular action by a correlation of the uterotropic activity and their ability to modulate the expression of estrogen-sensitive genes. We have analyzed the expression of androgen receptor (AR), progesterone receptor (PR), estrogen receptor (ER), clusterin (CLU), complement C3 (C3), and GAPDH mRNA in the rat uterus following oral administration of ethinylestradiol (EE), bisphenol A (BPA), o,p'-DDT (DDT), p-tert-octylphenol (OCT) and daidzein (DAI). A significant stimulation of the uterine wet weight could be observed after administration of all the substances. The activity of all analyzed compounds to stimulate uterine weight was low in comparison to EE. DDT has the highest activity to stimulate uterine weight whereas BPA and DAI turned out to be less potent. The analysis of gene expression revealed a very specific profile of molecular action in response to the different compounds which cannot be detected by judging the uterotropic response alone. A dose dependent analysis revealed that C3 mRNA is already modulated at doses where no uterotropic response was detectable. Although DAI and BPA were very weak stimulators of uterine growth, these substances were able to alter the expression of AR, ER and C3 very strongly. Based on these investigations the analyzed compounds can be subdivided into distinct classes: First, compounds which exhibit a similar gene expression fingerprint as EE (e.g. OCT); second, compounds exhibiting a significant uterotropic activity, but inducing a pattern of gene expression different from EE (e.g. DDT); and third, compounds like BPA and especially DAI which exhibit a very low uterotropic activity, but nevertheless modulate the expression of estrogen-sensitive genes. These findings strongly suggest that the fingerprint of uterine gene expression is a very sensitive tool to investigate estrogenicity of natural and synthetic compounds and offers the possibility to get information in regard to the molecular mechanisms involved in the action of the respective compounds.
Collapse
Affiliation(s)
- P Diel
- Institut für Morphologie und Tumorforschung, Deutsche Sporthochschule Köln, Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Lapointe J, Labrie C. Identification and cloning of a novel androgen-responsive gene, uridine diphosphoglucose dehydrogenase, in human breast cancer cells. Endocrinology 1999; 140:4486-93. [PMID: 10499502 DOI: 10.1210/endo.140.10.7071] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Androgens inhibit the growth of breast cancer cells, but the mechanism of androgen-induced growth inhibition has not yet been elucidated, and few androgen-responsive genes have been identified. We, therefore, used differential display PCR to identify novel androgen-responsive genes in ZR-75-1 human breast cancer cells. The human UDP-glucose dehydrogenase gene (UDPGDH), which was not known to be androgen regulated, was detected and cloned by complementary DNA library screening. The UDPGDH open reading frame codes for a protein of 494 amino acids that migrates at an apparent molecular mass of approximately 54 kDa. Northern blot analysis revealed the existence of two messenger RNA species of approximately 3.5 and 2.7 kb in all of the human breast cancer cell lines examined. The major UDPGDH transcript was induced rapidly (within 6 h) by dihydrotestosterone in ZR-75-1 cells, and a maximal 13-fold induction was observed after 24 h of treatment. The increase in UDPGDH messenger RNA was completely prevented by coincubation with the pure antiandrogen hydroxyflutamide, but not by cycloheximide, indicating that UDPGDH is directly regulated by the androgen receptor. As UDPGDH is required for the production of uridine 5'-diphosphoglucuronic acid, a substrate for the steroid-conjugating uridine diphospho-glucuronosyltransferase enzymes, up-regulation of UDPGDH expression by androgens might play an important role in the control of sex steroid inactivation via glucuronidation in breast cancer cells.
Collapse
Affiliation(s)
- J Lapointe
- Laboratory of Molecular Endocrinology, Laval University Medical Research Center and Laval University, Ste-Foy, Québec, Canada
| | | |
Collapse
|
44
|
Lapointe J, Fournier A, Richard V, Labrie C. Androgens down-regulate bcl-2 protooncogene expression in ZR-75-1 human breast cancer cells. Endocrinology 1999; 140:416-21. [PMID: 9886853 DOI: 10.1210/endo.140.1.6410] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although a large proportion of primary human breast cancers express the androgen receptor, and treatment with androgens exerts beneficial effects in women with breast cancer, the role and especially the mechanism of action of androgens in breast cancer development and growth are not well understood. The potential effect of androgens on bcl-2 protooncogene expression was investigated in a human breast cancer cell line whose proliferation is known to be inhibited by androgens. The estrogen-responsive ZR-75-1 cells were grown in the presence or absence of 5alpha-dihydrotestosterone (DHT), alone or in combination with 17beta-estradiol. DHT caused a marked down-regulation of Bcl-2 protein and messenger RNA levels in both the presence and absence of 17beta-estradiol. The inhibitory effect of DHT was completely prevented by coincubation with the pure antiandrogen hydroxyflutamide. The present data indicate that androgens can down-regulate bcl-2 protooncogene levels via an androgen receptor-mediated mechanism, thus providing a novel mechanism for their known inhibitory effect on breast cancer cell growth.
Collapse
Affiliation(s)
- J Lapointe
- Laboratory of Molecular Endocrinology, Centre Hospitalier de l'Université Laval Research Center and Laval University, Québec, Canada
| | | | | | | |
Collapse
|
45
|
Blasberg ME, Robinson S, Henderson LP, Clark AS. Inhibition of estrogen-induced sexual receptivity by androgens: role of the androgen receptor. Horm Behav 1998; 34:283-93. [PMID: 9878277 DOI: 10.1006/hbeh.1998.1484] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Both naturally occurring and synthetic androgens have been shown to inhibit estrogen-induced sexual receptivity when administered to ovariectomized (OVX) rats. The mechanisms by which androgens exert these effects, however, remain unclear. Experiments were conducted to determine the role of the androgen receptor in the inhibition of estrogen-induced sexual receptivity in OVX rats by using flutamide, an androgen receptor antagonist. In each experiment, OVX Long-Evans rats received 6 consecutive days of estradiol benzoate (EB; 2.0 microg/day) followed by 15 days of EB concurrent with flutamide (10. 0 mg/kg; twice daily) or the vehicle and one of the following androgens or the vehicle: dihydrotestosterone propionate (7.5 mg/kg), 3alpha-androstanediol (3.75 mg/kg), 17alpha-methyltestosterone (7.5 mg/kg), stanozolol (7.5 mg/kg), or nandrolone decanoate (7.5 mg/kg). On Day 15, all female rats received progesterone (P; 1.0 mg/rat) 4 h before testing. Tests for sexual receptivity were conducted on Days 3, 6, 14, and 15 of androgen/flutamide treatment. Each androgen inhibited sexual receptivity as expected, and concurrent treatment with flutamide reversed the inhibitory effects of all androgens on sexual receptivity on all test days. High levels of sexual receptivity were displayed in response to P on Day 15, regardless of experimental treatment. These results suggest that naturally occurring and synthetic androgens act at the androgen receptor to inhibit estrogen-induced sexual receptivity in OVX rats.
Collapse
Affiliation(s)
- M E Blasberg
- Department of Psychology, Dartmouth College, Hanover, New Hampshire 03755, USA
| | | | | | | |
Collapse
|
46
|
Beri R, Kumar N, Savage T, Benalcazar L, Sundaram K. Estrogenic and progestational activity of 7alpha-methyl-19-nortestosterone, a synthetic androgen. J Steroid Biochem Mol Biol 1998; 67:275-83. [PMID: 9879986 DOI: 10.1016/s0960-0760(98)00114-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthetic androgens exhibit estrogenic/antiestrogenic and progestational activities in addition to their androgenic effects. To investigate the pharmacological action of the synthetic androgen, 7alpha-methyl-19-nortestosterone (MENT), we examined its action in female rodents. The criteria employed for estrogenic/antiestrogenic effects were, uterine weight increase, vaginal cornification, induction of progesterone receptors (PR) synthesis and stimulation of peroxidase activity in the uteri of ovariectomized rats and mice. MENT increased uterine weight in a dose dependent manner, but did not cause vaginal cornification or stimulate PR synthesis in the uterus. The uterotropic activity of MENT was 200-fold lower than that of estradiol. Estrogen receptor (ER) bound [3H]-E2 was displaced by E2 and MENT with ED50 values of 70 pg and 250 ng, respectively, a 3,500 fold difference in their binding affinity. The low binding of MENT to ER, in contrast to its relatively high uterotropic action, suggested that receptors other than ER may be involved in its action on the uterus. The progestational activity of MENT in immature rabbits using the McPhail index assay was comparable to that of progesterone. Binding affinities of MENT and progesterone to PR were also comparable. However, the action of MENT on the uterus does not seem to be a progestational effect since mifepristone, an antiprogestin, had no effect on MENT-induced uterine growth. Specific androgen receptors (AR) in uterine cytosol were demonstrated. The involvement of AR in MENT action was confirmed by using an antiandrogen (flutamide) and an antiestrogen (ICI-182) in ovariectomized mice. Although MENT did not block the uterotropic effect of E2, it inhibited the E2-induced cornification of vaginal epithelium, induction of uterine PR synthesis and increase in uterine peroxidase activity in ovariectomized rats. The antiestrogenic effect of MENT was also blocked by flutamide. These results suggest that the uterotropic and antiestrogenic effects of androgens are mediated via AR. It is concluded that the increase in uterine weight caused by MENT is attributable to its anabolic effects.
Collapse
Affiliation(s)
- R Beri
- Center for Biomedical Research, The Population Council, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
47
|
Couillard S, Labrie C, Bélanger A, Candas B, Pouliot F, Labrie F. Effect of dehydroepiandrosterone and the antiestrogen EM-800 on growth of human ZR-75-1 breast cancer xenografts. J Natl Cancer Inst 1998; 90:772-8. [PMID: 9605648 DOI: 10.1093/jnci/90.10.772] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In the mammary gland, androgens are formed from the precursor steroid dehydroepiandrosterone (DHEA). Clinical evidence indicates that androgens have inhibitory effects on breast cancer. Estrogens, on the other hand, stimulate the development and growth of breast cancer. We studied the effect of DHEA alone or in combination with the newly described pure antiestrogen EM-800 on the growth of subcutaneous tumor xenografts formed by the human breast cancer cell line ZR-75-1 in ovariectomized nude mice. METHODS Immediately after ovariectomy, mice received daily subcutaneous injections of 0.5 microg estrone (E1) (an estrogenic hormone). EM-800 (15, 50, or 100 microg) was given orally once daily. DHEA was administered percutaneously twice daily (total dose of 0.3, 1.0, or 3.0 mg) to the dorsal skin either alone or in combination with a 15-microg daily oral dose of EM-800. Changes in tumor size in response to the treatments (in relation to measurements made on the first day of treatment) were assessed periodically. At the end of the experiments, tumors were dissected and weighed. RESULTS A 9.4-fold increase in tumor size in 9.5 months was observed in ovariectomized mice receiving E1 alone. Administration of 15, 50, or 100 microg EM-800 in E1-supplemented mice led to inhibitions of 87.5%, 93.5%, and 94.0% in tumor size, respectively. DHEA, on the other hand, at doses of 0.3, 1.0, or 3.0 mg inhibited terminal tumor size by 50.4%, 76.8%, and 80.0%, respectively. Comparable inhibitions in tumor size were obtained with a daily 15-microg oral dose of EM-800 with or without different doses of percutaneous DHEA. CONCLUSIONS DHEA and EM-800 independently suppressed the growth of E1-stimulated ZR-75-1 xenograft tumors in nude mice. Administration of DHEA at the defined doses did not alter the inhibitory effect of EM-800.
Collapse
Affiliation(s)
- S Couillard
- Laboratory of Molecular Endocrinology, CHUL Research Center and Laval University, Québec, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Although the androgen receptor (AR)3 is often co-expressed with the estrogen receptor (ER) and progesterone receptor (PR) in human breast tumors, its role in breast cancer is poorly understood. Specific growth stimulatory and inhibitory actions of androgens have been described in human breast cancer cell lines. The mechanisms by which androgens exert these contrasting growth effects are unknown. A commonly utilized second line therapy for the treatment of advanced breast cancer is high dose medroxyprogesterone acetate (MPA). Although MPA, a synthetic progestin, was thought to act exclusively through the PR, the androgenic side-effects observed in women taking MPA suggest that its action may also be mediated in part by the AR. In support of this hypothesis, the level of AR measured by radioligand binding in primary breast tumors was correlated with the duration of response to MPA treatment following failure of tamoxifen therapy. Recent data suggest that the presence of structurally altered AR in breast cancers may account for unresponsiveness to MPA in some of these cases. Further studies are warranted to determine the role of AR mediated pathways in regulating breast tumor growth. In particular, identification of androgen-regulated genes may lead to new possibilities for the hormonal treatment of breast cancer.
Collapse
Affiliation(s)
- S N Birrell
- Flinders Cancer Centre, School of Medicine, Flinders University of South Australia, Bedford Park, Australia
| | | | | |
Collapse
|
49
|
Abstract
Anabolic-androgenic steroid (AAS) compounds are synthetic androgens taken by athletes to increase physical strength and endurance. Recent studies in our laboratory have demonstrated that AAS administration disrupts the estrous cycle of Long-Evans rats. The present experiments examined the effects of six commonly abused AAS compounds on sexual receptivity in ovariectomized rats. Adult female Long-Evans rats received estradiol benzoate (EB; 2.0 micrograms/day s.c.) for 6 consecutive days followed by 15 days of EB concurrent with daily s.c. injections of 7.5 mg/kg of one of the following AAS compounds: 17 alpha-methyltestosterone, methandrostenolone, nandrolone decanoate, stanozolol, oxymetholone, testosterone cypionate, or the oil vehicle. On Day 15, all female rats received progesterone (1.0 mg/rat) 4 h before testing. Tests for sexual receptivity were conducted on Days 3, 6, 14, and 15 of AAS treatment. Although the time course of AAS effects on sexual receptivity varied, some overall effects were clear. For example, 17 alpha-methyltestosterone, methandrostenolone, nandrolone decanoate, and stanozolol interfered with the display of sexual receptivity on Day 14, whereas oxymetholone and testosterone cypionate had no effect. Rats in all groups displayed high levels of sexual receptivity after receiving progesterone on Day 15. Our results show that AAS compounds vary in their degree of inhibition of female sexual behavior in ovariectomized rats.
Collapse
Affiliation(s)
- M E Blasberg
- Department of Psychology, Dartmouth College, Hanover, New Hampshire 03755, USA
| | | |
Collapse
|
50
|
Affiliation(s)
- G Boccuzzi
- Department of Clinical Pathophysiology, University of Turin, Italy
| | | |
Collapse
|