1
|
Important roles of estrogen receptor alpha in tumor progression and anti-estrogen therapy of pancreatic ductal adenocarcinoma. Life Sci 2020; 260:118302. [PMID: 32827543 DOI: 10.1016/j.lfs.2020.118302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
AIMS The roles of estrogen receptors (ERs) and the efficacy of anti-estrogen (E2) therapies in pancreatic cancer stay controversial. The main objectives of this study were to investigate the potential roles of ERs in tumor progression and endocrine therapies. MAIN METHODS The ER expression status in PANC-1 and SW1990 pancreatic cancer cell lines was determined. SRB assay, colony formation assay and proliferation assay were used to investigate the responses of these cells to E2. ERα-selective agonist propylpyrazoletriol (PPT), ERβ-selective agonist diarylpropionitrile (DPN), ERα over-expressed SW1990 cells, ERα knock-out PANC-1 cells and patient-derived xenografts (PDX) were applied to investigate the potential roles of ERα in pancreatic cancer. The phosphorylation of ERα-related signaling molecules extracellular regulated protein kinases (ERK1/2) and protein kinase B (AKT) were investigated. The in vivo anti-tumor efficacy and safety of letrozole (LTZ) combined with leuprorelin acetate (LA) and gemcitabine (GEM) were also preliminarily studied. KEY FINDINGS PANC-1 cells expressed much more ERα than SW1990 cells, and ERβ level was with less diversity. Accordingly, the proliferation of PANC-1 rather than SW1990 cells could be stimulated by E2, and only PANC-1 could respond to LTZ endocrine therapy in female but not male mice. The phosphorylation of ERK1/2 but not AKT was altered by over-expressed or knocking out of ERα with or without the addition of E2 and LTZ. The combination therapy of LTZ and GEM showed acceptable efficacy and safety. SIGNIFICANCE This study showed the important roles of ERα in tumor progression and endocrine therapies of pancreatic cancer in women.
Collapse
|
2
|
Yancu D, Viau R, Sanderson T. Development of an estrogen-dependent breast cancer co-culture model as a tool for studying endocrine disruptors. Toxicol In Vitro 2019; 62:104658. [PMID: 31629071 DOI: 10.1016/j.tiv.2019.104658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/14/2019] [Accepted: 09/17/2019] [Indexed: 10/25/2022]
Abstract
We developed an innovative co-culture system composed of Hs578t human mammary stromal-like cells and T47D hormone-dependent breast epithelial tumor cells as a representative in vitro model of the human hormone-dependent mammary tumor microenvironment. Hs578t cells expressed aromatase (CYP19) mainly via the healthy stromal CYP19 promoter I.4, but also to a lesser extent via the breast cancer-relevant promoters PII, I.3 and I.7, and produced estrogens from androgen precursors. These estrogens stimulated T47D cell proliferation and estrogen receptor-dependent expression of trefoil factor-1 (TFF1), which is known to stimulate mammary tumor cell proliferation and migration. Hs578t cells can also undergo a "promoter-switch" where the normally silent CYP19 promoters PII, I.3 and I.7 become activated, which mimics the in vivo situation in human breast cancer patients. This positive feedback loop is the hallmark of the hormone-dependent breast tumor microenvironment. Using the co-culture model we designed, we evaluated the promoter-specific expression of CYP19, expression of estrogen-dependent gene TFF1, and determined the effects exhibited by basil and fennel seed essential oils on steroidogenesis in the tumor microenvironment.
Collapse
Affiliation(s)
- Debbie Yancu
- INRS - Institut Armand-Frappier, Laval, QC H7V 1B7, Canada.
| | - Rachel Viau
- INRS - Institut Armand-Frappier, Laval, QC H7V 1B7, Canada
| | | |
Collapse
|
3
|
Sang X, Han H, Li T, Lin SX. Mutual regulations and breast cancer cell control by steroidogenic enzymes: Dual sex-hormone receptor modulation upon 17β-HSD7 inhibition. J Steroid Biochem Mol Biol 2019; 193:105411. [PMID: 31207361 DOI: 10.1016/j.jsbmb.2019.105411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/28/2019] [Accepted: 06/13/2019] [Indexed: 12/23/2022]
Abstract
Reductive 17β-hydroxysteroid dehydrogenases (17β-HSDs) and 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) play crucial roles in respectively regulating steroids and glucocorticoids for the progression of hormone-dependent breast cancer. Most studies focused on the function and individual regulation of these enzymes. However, mutual regulation of these enzymes and the induced modulation on the estrogen and androgen receptors for breast cancer promotion are not yet clear. In this study, MCF-7 and T47D cells were treated with inhibitors of 17β-HSD1, 17β-HSD7, aromatase or steroid sulfatase (STS), then mRNA levels of 17β-HSD7, STS, 11β-HSD 2, estrogen receptors α (ERα) and androgen receptor (AR) were determined by Q-PCR. ER negative cell line MDA-MB-231 was used as a negative control. Our results demonstrate that 17β-HSD7, STS and 11β-HSD2 are all regulated by the same estrogen estradiol via ERα. When the gene of ERα (ESR1) was knocked down, there was no longer significant mutual regulation of these enzymes. Our results demonstrate that important steroidogenic enzymes transcriptionally regulated by ERα, can be mutually closely correlated. Inhibition of one of them can reduce the expression of another, thereby amplifying the role of the inhibition. Furthermore, inhibition of 17β-HSD7 increases the expression of AR gene which is considered as a marker for better prognosis in ER + breast cancer, while maintaining ERα level. Thus, our mechanistic finding provides a base for further improving the endocrine therapy of ER + breast cancer, e.g., for selecting the target steroid enzymes, and for the combined targeting of human 17β-HSD7 and ERα.
Collapse
Affiliation(s)
- Xiaoye Sang
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec G1V4G2, Canada
| | - Hui Han
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec G1V4G2, Canada; Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Tang Li
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec G1V4G2, Canada
| | - Sheng-Xiang Lin
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec G1V4G2, Canada.
| |
Collapse
|
4
|
Xiao B, Wen J, Zhao C, Chen L, Sun Z, Li L. [Differences in expression profiles of circular RNA between luminal breast cancer cells and normal breast cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 38:1014-1019. [PMID: 30187868 DOI: 10.3969/j.issn.1673-4254.2018.08.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the differences in the expression profiles of circular RNA (circRNA) between luminal breast cancer cells and normal breast cells. METHODS Total RNA extracted from luminal breast cancer cells MCF7 and normal breast cells MCF10A was digested with Rnase R to remove linear RNAs and enrich circRNAs. The enriched circRNAs were amplified and transcribed into fluorescent cRNAs using a random priming method, and were hybridized onto the circRNA hybridization array. The circRNA expression profiles of MCF7 and MCF10A cells were analyzed using Agilent Feature Extraction software. Quantile normalization and subsequent data processing were performed, and volcano plot filtering and hierarchical clustering were utilized to analyze the circRNA expression patterns. The expressions of 3 circRNAs with significant log fold changes were validated using qPCR. RESULTS The hybridization array data revealed significant differences in the circRNA expression profiles between MCF7 and MCF10A cells. Compared with those of MCF10A cells, the 12910 circRNAs expressed in MCF7 cells showed 5964 up-regulated, 81 consistently regulated, and 6865 down-regulated circRNAs; 343 circRNAs showed a log fold change by more than 2 folds, among which 213 circRNAs were up-regulated and 130 were down-regulated. Nine circRNAs showed differential expressions by more than 2 folds, including 8 up-regulated ones, namely hsa_circRNA_061260 (6.02 folds), hsa_circRNA_103933 (5.96 folds), hsa_circRNA_005239 (5.84 folds), hsa_circRNA_100689 (5.69 folds), hsa_circRNA_004087 (5.60 folds), hsa_circRNA_104420 (5.25 folds), hsa_circRNA_104421 (5.13 folds) and hsa_circRNA_101222 (5.03 folds); only one circRNA was down-regulated, namely hsa_circRNA_104864 (5.09 folds). The expressions of hsa_circRNA_100689, hsa_circRNA_005239 and hsa_circRNA_104864 were further validated by qPCR, which yielded consistent results with the microarray data. CONCLUSIONS The circRNA expression profiles differ significantly between luminal breast cancer cells and normal breast cells. These differentially expressed circRNAs may serve as potential novel targets for the diagnosis of luminal breast cancer.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Laboratory Medicine, Guangzhou General Hospital of PLA, Guangzhou 510010, China
| | - Jiaxin Wen
- Department of Medical Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 501515, China
| | - Chaoran Zhao
- Department of Medical Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou 501515, China
| | - Lidan Chen
- Department of Laboratory Medicine, Guangzhou General Hospital of PLA, Guangzhou 510010, China
| | - Zhaohui Sun
- Department of Laboratory Medicine, Guangzhou General Hospital of PLA, Guangzhou 510010, China
| | - Linhai Li
- Department of Laboratory Medicine, Guangzhou General Hospital of PLA, Guangzhou 510010, China
| |
Collapse
|
5
|
Yang Y, Li J, Song Q, Zhu K, Yu X, Tian Y, Zhang J. Reduction in milk fat globule-EGF factor 8 inhibits triple-negative breast cancer cell viability and migration. Oncol Lett 2019; 17:3457-3465. [PMID: 30867784 PMCID: PMC6396170 DOI: 10.3892/ol.2019.9968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 01/29/2018] [Indexed: 11/06/2022] Open
Abstract
Milk fat globule-EGF factor 8 (MFG-E8) has been demonstrated to be associated with the progression and metastasis of breast cancer, although the underlying mechanisms remain undefined. The aim of the present study was to explore the role of MFG-E8 in human breast cancer and examine the underlying molecular mechanisms. Reverse transcription-quantitative polymerase chain reaction analysis was used to evaluate the expression levels of MFG-E8 in human breast carcinoma cell lines. An MFG-E8 small interfering RNA lentiviral vector was constructed and transfected into MDA-MB-231 cells. The results indicated that the in vitro silencing of MFG-E8 significantly inhibited the viability, invasion and migration of breast cancer cells. By using a flow cytometric approach, the knockdown of MFG-E8 was revealed to significantly induce cell cycle arrest at the G2/M phase and cell apoptosis. Furthermore, the downregulation of MFG-E8 induced the activation of apoptosis-associated proteins, and inhibited the expression of matrix metalloproteinase and epithelial-mesenchymal transition-associated proteins. Collectively, the results of the present study emphasize the importance of MFG-E8 deregulation in mammary carcinogenesis and its potential use as a biomarker for the diagnosis of breast carcinomas.
Collapse
Affiliation(s)
- Yong Yang
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jiebao Li
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qi Song
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kongjun Zhu
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaocheng Yu
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ye Tian
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jiaheng Zhang
- Department of Breast and Thyroid Surgery, Wuhan No. 1 Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
6
|
Sang X, Han H, Poirier D, Lin SX. Steroid sulfatase inhibition success and limitation in breast cancer clinical assays: An underlying mechanism. J Steroid Biochem Mol Biol 2018; 183:80-93. [PMID: 29803725 DOI: 10.1016/j.jsbmb.2018.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/18/2018] [Accepted: 05/23/2018] [Indexed: 12/23/2022]
Abstract
Steroid sulfatase is detectable in most hormone-dependent breast cancers. STX64, an STS inhibitor, induced tumor reduction in animal assay. Despite success in phase І clinical trial, the results of phase II trial were not that significant. Breast Cancer epithelial cells (MCF-7 and T47D) were treated with two STS inhibitors (STX64 and EM1913). Cell proliferation, cell cycle, and the concentrations of estradiol and 5α-dihydrotestosterone were measured to determine the endocrinological mechanism of sulfatase inhibition. Comparisons were made with inhibitions of reductive 17β-hydroxysteroid dehydrogenases (17β-HSDs). Proliferation studies showed that DNA synthesis in cancer cells was modestly decreased (approximately 20%), accompanied by an up to 6.5% in cells in the G0/G1 phase and cyclin D1 expression reduction. The concentrations of estradiol and 5α-dihydrotestosterone were decreased by 26% and 3% respectively. However, supplementation of 5α-dihydrotestosterone produced a significant increase (approximately 35.6%) in the anti-proliferative effect of sulfatase inhibition. This study has clarified sex-hormone control by sulfatase in BC, suggesting that the different roles of estradiol and 5α-dihydrotestosterone can lead to a reduction in the effect of sulfatase inhibition when compared with 17β-HSD7 inhibition. This suggests that combined treatment of sulfatase inhibitors with 17β-HSD inhibitors such as the type7 inhibitor could hold promise for hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Xiaoye Sang
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada
| | - Hui Han
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada; Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Donald Poirier
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada
| | - Sheng-Xiang Lin
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada.
| |
Collapse
|
7
|
Rubinow KB. An intracrine view of sex steroids, immunity, and metabolic regulation. Mol Metab 2018; 15:92-103. [PMID: 29551633 PMCID: PMC6066741 DOI: 10.1016/j.molmet.2018.03.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/26/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
Abstract
Background Over the past two decades, parallel recognition has grown of the importance of both sex steroids and immune activity in metabolic regulation. More recently, these discrete areas have been integrated in studies examining the metabolic effects of sex steroid immunomodulation. Implicit in these studies has been a traditional, endocrine model of sex steroid delivery from the gonads to target cells, including immune cells. Thus, research to date has focused on the metabolic effects of sex steroid receptor signaling in immune cells. This endocrine model, however, overlooks the extensive capacity of immune cells to generate and metabolize sex steroids, enabling the production of sex steroids for intracrine signaling – that is, sex steroid production for signaling within the cell of origin. Intracrine function allows highly cell-autonomous regulation of sex steroid exposure, and sex steroid secretion by immune cells could confer paracrine signaling effects in neighboring cells within metabolic tissues. In this review, immune cell intracrinology will denote sex steroid production within immune cells for either intracrine or paracrine signaling. This intracrine capacity of immune cells has been well established, and prior work has supported its importance in autoimmune disorders, trauma, and cancer. The potential relevance of immune cell intracrine function to the regulation of energy balance, body weight, body composition, and insulin sensitivity has yet to be explored. Scope of review The following review will detail findings to date regarding the steroidogenic and steroid metabolizing capacity of immune cells, the regulation of immune cell intracrine function, and the biological effects of immune-derived sex steroids, including the clinical relevance of immune cell intracrinology in fields other than metabolism. These findings will serve as the basis for a proposed model of immune cell intracrinology constituting a new frontier in metabolism research. Major conclusions The development of highly sensitive mass spectrometric methods for sex steroid measurement and quantitation of metabolic flux now allows unprecedented ability to interrogate sex steroid production, metabolism and secretion by immune cells. Immune cell intracrinology could reveal key mechanisms underlying immune cell-mediated metabolic regulation. Sex steroids exert immunomodulatory effects that may influence metabolic health. Immune cells can synthesize, modify, and metabolize sex steroids. Immune cell-derived sex steroids may play intracrine, autocrine, paracrine, and possibly even endocrine roles. Immune cell steroidogenesis is a largely unexplored area of metabolism research.
Collapse
Affiliation(s)
- Katya B Rubinow
- Diabetes Institute, Department of Medicine, University of Washington, School of Medicine, 850 Republican St., Box 358055, Seattle, WA 98109, USA.
| |
Collapse
|
8
|
Kikuchi K, McNamara KM, Miki Y, Moon JY, Choi MH, Omata F, Sakurai M, Onodera Y, Rai Y, Ohi Y, Sagara Y, Miyashita M, Ishida T, Ohuchi N, Sasano H. Effects of cytokines derived from cancer-associated fibroblasts on androgen synthetic enzymes in estrogen receptor-negative breast carcinoma. Breast Cancer Res Treat 2017; 166:709-723. [PMID: 28831645 DOI: 10.1007/s10549-017-4464-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/12/2017] [Indexed: 02/03/2023]
Abstract
PURPOSE The tumor microenvironment plays pivotal roles in promotion of many malignancies. Cancer-associated fibroblasts (CAFs) have been well-known to promote proliferation, angiogenesis, and metastasis but mechanistic understanding of tumor-stroma interactions is not yet complete. Recently, estrogen synthetic enzymes were reported to be upregulated by co-culture with stromal cells in ER positive breast carcinoma (BC) but effects of co-culture on androgen metabolism have not been extensively examined. Therefore, we evaluated roles of CAFs on androgen metabolism in ER-negative AR-positive BC through co-culture with CAFs. METHODS Concentrations of steroid hormone in supernatant of co-culture of MDA-MB-453 and primary CAFs were measured using GC-MS. Cytokines derived from CAFs were determined using Cytokine Array. Expressions of androgen synthetic enzymes were confirmed using RT-PCR and Western blotting. Correlations between CAFs and androgen synthetic enzymes were analyzed using triple-negative BC (TNBC) patient tissues by immunohistochemistry. RESULTS CAFs were demonstrated to increase expressions and activities of 17βHSD2, 17βHSD5, and 5α-Reductase1. IL-6 and HGF that were selected as potential paracrine mediators using cytokine array induced 17βHSD2, 17βHSD5, and 5α-Reductase1 expression. Underlying mechanisms of IL-6 paracrine regulation of 17βHSD2 and 17βHSD5 could be partially dependent on phosphorylated STAT3, while phosphorylated ERK could be involved in HGF-mediated 5α-Reductase1 induction. α-SMA status was also demonstrated to be significantly correlated with 17βHSD2 and 17βHSD5 status in TNBC tissues, especially AR-positive cases. CONCLUSIONS Results of our present study suggest that both IL-6 and HGF derived from CAFs could contribute to the intratumoral androgen metabolism in ER-negative BC patients.
Collapse
Affiliation(s)
- Kyoko Kikuchi
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Keely May McNamara
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan.
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science (IRIDeS), Tohoku University, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Ju-Yeon Moon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seoul, 02792, Korea
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seoul, 02792, Korea
| | - Fumiya Omata
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Minako Sakurai
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Yoshiaki Onodera
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Yoshiaki Rai
- Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima, Japan
| | - Yasuyo Ohi
- Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima, Japan
| | - Yasuaki Sagara
- Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Noriaki Ohuchi
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-Ku, Sendai-shi, Miyagi, 980-8575, Japan
| |
Collapse
|
9
|
Chanawong A, Mackenzie PI, McKinnon RA, Hu DG, Meech R. Exemestane and Its Active Metabolite 17-Hydroexemestane Induce UDP-Glucuronosyltransferase (UGT) 2B17 Expression in Breast Cancer Cells. J Pharmacol Exp Ther 2017; 361:482-491. [PMID: 28404691 DOI: 10.1124/jpet.117.240317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/06/2017] [Indexed: 11/22/2022] Open
Abstract
Exemestane (EXE) is an aromatase inhibitor indicated for endocrine therapy of breast cancer in postmenopausal women. The primary active metabolite of EXE, 17-hydroexemestane (17-HE), is inactivated via glucuronidation, mainly by UDP-glucuronosyltransferase 2B17 (UGT2B17). UGT2B17 also has a primary role in inactivation of endogenous androgens testosterone and dihydrotestosterone and may play an important role in regulation of breast and prostate tumor intracrinology. We recently reported that UGT2B17 could be induced by both estrogenic and androgenic ligands in breast cancer cells via binding of the estrogen receptor α (ERα) or the androgen receptor (AR) to a complex regulatory unit in the proximal UGT2B17 promoter. In this study we show that both EXE and 17-HE increase UGT2B17 mRNA levels in breast cancer MCF-7 and MDA-MB-453 cells, and increase glucuronidation of UGT2B17 substrates, including 17-HE and androsterone. Using antagonists of ERα and AR as well as inhibition mediated by small interfering RNA (siRNA) we demonstrate that EXE and 17-HE induce UGT2B17 expression primarily via the AR. This result is consistent with previous reports that 17-HE can act as an AR ligand. In vitro studies suggest that multiple steroid-responsive DNA elements within the proximal promoter are involved in the response to 17-HE-liganded AR. The up-regulation of UGT2B17 by EXE and 17-HE in breast cancer cells might enhance the local metabolism of 17-HE as well as that of endogenous androgens, hence impacting potentially on treatment outcomes.
Collapse
Affiliation(s)
- Apichaya Chanawong
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University School of Medicine, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|