1
|
Yuan J, Yang L, Li Z, Zhang H, Wang Q, Wang B, Chinnathambi A, Govindasamy C, Basappa S, Nagaraja O, Madegowda M, Beeraka NM, Nikolenko VN, Wang M, Wang G, Rangappa KS, Basappa B. Pyrimidine-triazole-tethered tert-butyl-piperazine-carboxylate suppresses breast cancer by targeting estrogen receptor signaling and β-catenin activation. IUBMB Life 2024. [PMID: 39275910 DOI: 10.1002/iub.2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/16/2024]
Abstract
Several chemotherapeutics against breast cancer are constrained by their adverse effects and chemoresistance. The development of novel chemotherapeutics to target metastatic breast cancer can bring effective clinical outcomes. Many breast cancer patients present with tumors that are positive for estrogen receptors (ERs), highlighting the importance of targeting the ER pathway in this particular subtype. Although selective estrogen receptor modulators (SERMs) are commonly used, their side effects and resistance issues necessitate the development of new ER-targeting agents. In this study, we report that a newly synthesized compound, TTP-5, a hybrid of pyrimidine, triazole, and tert-butyl-piperazine-carboxylate, effectively binds to estrogen receptor alpha (ERα) and suppresses breast cancer cell growth. We assessed the impact of TTP-5 on cell proliferation using MTT and colony formation assays and evaluated its effect on cell motility through wound healing and invasion assays. We further explored the mechanism of action of this novel compound by detecting protein expression changes using Western blotting. Molecular docking was used to confirm the interaction of TTP-5 with ERα. The results indicated that TTP-5 significantly reduced the proliferation of MCF-7 cells by blocking the ERα signaling pathway. Conversely, although it did not influence the growth of MDA-MB-231 cells, TTP-5 hindered their motility by modulating the expression of proteins associated with epithelial-mesenchymal transition (EMT), possibly via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhi Li
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Hua Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qun Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Bei Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Shreeja Basappa
- Department of Chemistry, BITS-Pilani, Hyderabad Campus, Medchal, India
| | | | | | - Narasimha M Beeraka
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, India
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Minghua Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Geng Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | | | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, India
| |
Collapse
|
2
|
Baker R, Dell'Acqua G, Richards A, Thornton MJ. Nutraceuticals known to promote hair growth do not interfere with the inhibitory action of tamoxifen in MCF7, T47D and BT483 breast cancer cell lines. PLoS One 2024; 19:e0297080. [PMID: 38408073 PMCID: PMC10896530 DOI: 10.1371/journal.pone.0297080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/26/2023] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Hair loss/thinning is a common side effect of tamoxifen in estrogen receptor (ER) positive breast cancer therapy. Some nutraceuticals known to promote hair growth are avoided during breast cancer therapy for fear of phytoestrogenic activity. However, not all botanical ingredients have similarities to estrogens, and in fact, no information exists as to the true interaction of these ingredients with tamoxifen. Therefore, this study sought to ascertain the effect of nutraceuticals (+/- estrogen/tamoxifen), on proliferation of breast cancer cells and the relative expression of ERα/β. METHODS Kelp, Astaxanthin, Saw Palmetto, Tocotrienols, Maca, Horsetail, Resveratrol, Curcumin and Ashwagandha were assessed on proliferation of MCF7, T47D and BT483 breast cancer cell lines +/- 17β-estradiol and tamoxifen. Each extract was analysed by high performance liquid chromatography (HPLC) prior to use. Cellular ERα and ERβ expression was assessed by qRT-PCR and western blot. Changes in the cellular localisation of ERα:ERβ and their ratio following incubation with the nutraceuticals was confirmed by immunocytochemistry. RESULTS Estradiol stimulated DNA synthesis in three different breast cancer cell lines: MCF7, T47D and BT483, which was inhibited by tamoxifen; this was mirrored by a specific ERa agonist in T47D and BT483 cells. Overall, nutraceuticals did not interfere with tamoxifen inhibition of estrogen; some even induced further inhibition when combined with tamoxifen. The ERα:ERβ ratio was higher at mRNA and protein level in all cell lines. However, incubation with nutraceuticals induced a shift to higher ERβ expression and a localization of ERs around the nuclear periphery. CONCLUSIONS As ERα is the key driver of estrogen-dependent breast cancer, if nutraceuticals have a higher affinity for ERβ they may offer a protective effect, particularly if they synergize and augment the actions of tamoxifen. Since ERβ is the predominant ER in the hair follicle, further studies confirming whether nutraceuticals can shift the ratio towards ERβ in hair follicle cells would support a role for them in hair growth. Although more research is needed to assess safety and efficacy, this promising data suggests the potential of nutraceuticals as adjuvant therapy for hair loss in breast cancer patients receiving endocrine therapy.
Collapse
Affiliation(s)
- Richard Baker
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | | | | | - M Julie Thornton
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| |
Collapse
|
3
|
Miziak P, Baran M, Błaszczak E, Przybyszewska-Podstawka A, Kałafut J, Smok-Kalwat J, Dmoszyńska-Graniczka M, Kiełbus M, Stepulak A. Estrogen Receptor Signaling in Breast Cancer. Cancers (Basel) 2023; 15:4689. [PMID: 37835383 PMCID: PMC10572081 DOI: 10.3390/cancers15194689] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogen receptor (ER) signaling is a critical regulator of cell proliferation, differentiation, and survival in breast cancer (BC) and other hormone-sensitive cancers. In this review, we explore the mechanism of ER-dependent downstream signaling in BC and the role of estrogens as growth factors necessary for cancer invasion and dissemination. The significance of the clinical implications of ER signaling in BC, including the potential of endocrine therapies that target estrogens' synthesis and ER-dependent signal transmission, such as aromatase inhibitors or selective estrogen receptor modulators, is discussed. As a consequence, the challenges associated with the resistance to these therapies resulting from acquired ER mutations and potential strategies to overcome them are the critical point for the new treatment strategies' development.
Collapse
Affiliation(s)
- Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Marzena Baran
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Alicja Przybyszewska-Podstawka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland;
| | - Magdalena Dmoszyńska-Graniczka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| |
Collapse
|
4
|
Revisiting the Syndecans: Master Signaling Regulators with Prognostic and Targetable Therapeutic Values in Breast Carcinoma. Cancers (Basel) 2023; 15:cancers15061794. [PMID: 36980680 PMCID: PMC10046401 DOI: 10.3390/cancers15061794] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Syndecans (SDC1 to 4), a family of cell surface heparan sulfate proteoglycans, are frequently expressed in mammalian tissues. SDCs are aberrantly expressed either on tumor or stromal cells, influencing cancer initiation and progression through their pleiotropic role in different signaling pathways relevant to proliferation, cell-matrix adhesion, migration, invasion, metastasis, cancer stemness, and angiogenesis. In this review, we discuss the key roles of SDCs in the pathogenesis of breast cancer, the most common malignancy in females worldwide, focusing on the prognostic significance and molecular regulators of SDC expression and localization in either breast tumor tissue or its microenvironmental cells and the SDC-dependent epithelial–mesenchymal transition program. This review also highlights the molecular mechanisms underlying the roles of SDCs in regulating breast cancer cell behavior via modulation of nuclear hormone receptor signaling, microRNA expression, and exosome biogenesis and functions, as well as summarizing the potential of SDCs as promising candidate targets for therapeutic strategies against breast cancer.
Collapse
|
5
|
Olayoku FR, Verhoog NJD, Louw A. Cyclopia extracts act as selective estrogen receptor subtype downregulators in estrogen receptor positive breast cancer cell lines: Comparison to standard of care breast cancer endocrine therapies and a selective estrogen receptor agonist and antagonist. Front Pharmacol 2023; 14:1122031. [PMID: 36992834 PMCID: PMC10040842 DOI: 10.3389/fphar.2023.1122031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
Breast cancer is the most diagnosed type of cancer amongst women in economically developing countries and globally. Most breast cancers express estrogen receptor alpha (ERα) and are categorized as positive (ER+) breast cancer. Endocrine therapies such as, selective estrogen receptor modulators (SERMs), aromatase inhibitors (AIs), and selective estrogen receptor downregulators (SERDs) are used to treat ER+ breast cancer. However, despite their effectiveness, severe side-effects and resistance are associated with these endocrine therapies. Thus, it would be highly beneficial to develop breast cancer drugs that are as effective as current therapies, but less toxic with fewer side effects, and less likely to induce resistance. Extracts of Cyclopia species, an indigenous South African fynbos plant, have been shown to possess phenolic compounds that exhibit phytoestrogenic and chemopreventive activities against breast cancer development and progression. In the current study, three well characterized Cyclopia extracts, SM6Met, cup of tea (CoT) and P104, were examined for their abilities to modulate the levels of the estrogen receptor subtypes, estrogen receptor alpha and estrogen receptor beta (ERβ), which have been recognized as crucial to breast cancer prognosis and treatment. We showed that the Cyclopia subternata Vogel (C. subternata Vogel) extracts, SM6Met and cup of tea, but not the C. genistoides extract, P104, reduced estrogen receptor alpha protein levels while elevating estrogen receptor beta protein levels, thereby reducing the ERα:ERβ ratio in a similar manner as standard of care breast cancer endocrine therapies such as fulvestrant (selective estrogen receptor downregulator) and 4-hydroxytamoxifen (elective estrogen receptor modulator). Estrogen receptor alpha expression enhances the proliferation of breast cancer cells while estrogen receptor beta inhibits the proliferative activities of estrogen receptor alpha. We also showed that in terms of the molecular mechanisms involved all the Cyclopia extracts regulated estrogen receptor alpha and estrogen receptor beta protein levels through both transcriptional and translational, and proteasomal degradation mechanisms. Therefore, from our findings, we proffer that the C. subternata Vogel extracts, SM6Met and cup of tea, but not the C. genistoides extract, P104, selectively modulate estrogen receptor subtypes levels in a manner that generally supports inhibition of breast cancer proliferation, thereby demonstrating attributes that could be explored as potential therapeutic agents for breast cancer.
Collapse
|
6
|
Li Y, Jia Y, Wang X, Shang H, Tian Y. Protein-Targeted Degradation Agents Based on Natural Products. Pharmaceuticals (Basel) 2022; 16:ph16010046. [PMID: 36678543 PMCID: PMC9865760 DOI: 10.3390/ph16010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/17/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022] Open
Abstract
Natural products are an important source of drug lead compounds, and natural products with significant biological activity are constantly being discovered and used in clinical practice. At present, natural products play an important role in the targeted therapy of cancer, cardiovascular and cerebrovascular diseases, nervous system diseases, and autoimmune diseases. Meanwhile, in recent years, the rise of protein-targeted degradation technologies, such as proteolysis-targeting chimeras (PROTACs) and molecular glues, has provided a new solution for drug resistance caused by clinical molecular-targeting drugs. It is noteworthy that natural products and their derivatives, as important components of PROTACs and molecular glues, play an important role in the development of protein-targeting drugs. Hence, this review summarized the protein-targeted degradation agents based on natural products, such as PROTACs and molecular glues. More natural products with the potential to be used in the development of PROTACs and molecular glues as targeted protein degradation agents are still being investigated.
Collapse
Affiliation(s)
| | | | | | | | - Yu Tian
- Correspondence: (H.S.); (Y.T.)
| |
Collapse
|
7
|
Perrone E, Tudisco R, Pafundi PC, Guido D, Ciucci A, Martinelli E, Zannoni GF, Piermattei A, Spadola S, Ferrante G, Marchetti C, Scambia G, Fagotti A, Gallo D. What’s beyond BRCA Mutational Status in High Grade Serous Ovarian Cancer? The Impact of Hormone Receptor Expression in a Large BRCA-Profiled Ovarian Cancer Patient Series: A Retrospective Cohort Study. Cancers (Basel) 2022; 14:cancers14194588. [PMID: 36230510 PMCID: PMC9559459 DOI: 10.3390/cancers14194588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/17/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Ovarian hormones are involved in ovarian cancer pathogenesis. However, few reports have investigated the hormone receptor pattern according to BRCA mutational status. The aim of this single-center, observational, retrospective study was to explore the relationship between hormone receptor status and BRCA1/2 mutation in a cohort of 207 high-grade serous ovarian carcinoma (HGSOC) patients. Interesting differences emerged between BRCA-mutated and BRCA wild-type women, in terms of pattern of receptor expression and its association to the outcome. On the whole, our findings, though needing further validation, extend our understanding of the complex interplay between BRCA1/2 protein and hormone signaling, suggesting new pathways to be exploited in order to develop future personalized therapy. Abstract Several studies have explored the prognostic role of hormone receptor status in high-grade serous ovarian cancer (HGSOC) patients. However, few reports have investigated their expression according to BRCA mutational status. The aim of this single-center, observational, retrospective study was to explore the hormone receptor pattern and its potential prognostic role in a cohort of 207 HGSOC women stratified for BRCA mutational status. To this end, ERα, ERβ1, ERβ2, ERβ5, PR, and AR expression were assessed by immunohistochemistry in 135 BRCA-wild type (BRCA-wt) and 72 BRCA1/2 mutation carriers (BRCA-mut). No significant difference emerged in hormone receptor expression between the two sub-samples, except for a significantly lower ERα expression observed in pre-menopausal BRCA1/2-mut as compared to BRCA-wt patients (p = 0.02). None of the examined hormone receptors has revealed a significant prognostic role in the whole sample, apart from the ratio ERα/ERβ5 nuclear, for which higher values disclosed a positive role on the outcome in BRCA-wt subgroup (HR 0.77; CI 0.61–0.96; p = 0.019). Conversely, it negatively affected overall survival in the presence of BRCA1/2-mut (HR 1.41; CI 1.06–1.87; p = 0.020). Finally, higher PR levels were associated with platinum sensitivity in the whole sample (p = 0.019). Our data, though needing further validation, suggest a potential role of oestrogen-mediated pathways in BRCA1/2-associated HGSOC tumorigenesis, thus revealing a possible therapeutic potential for targeting this interaction.
Collapse
Affiliation(s)
- Emanuele Perrone
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Riccardo Tudisco
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Pia Clara Pafundi
- Epidemiology and Biostatistics Facility Core Research, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Davide Guido
- Bioinformatics Facility Core Research, Gemelli Science and Technology Park (GSTeP) Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandra Ciucci
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Unit of Translational Medicine for Woman and Child Health, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Enrica Martinelli
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Unit of Translational Medicine for Woman and Child Health, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Gian Franco Zannoni
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Gynecopathology and Breast Pathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessia Piermattei
- Gynecopathology and Breast Pathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Saveria Spadola
- Gynecopathology and Breast Pathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giulia Ferrante
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Claudia Marchetti
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Anna Fagotti
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Daniela Gallo
- Universita’ Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Unit of Translational Medicine for Woman and Child Health, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Francesco Vito 1, 00168 Rome, Italy
- Correspondence:
| |
Collapse
|
8
|
Velesiotis C, Kanellakis M, Vynios DH. Steviol glycosides affect functional properties and macromolecular expression of breast cancer cells. IUBMB Life 2022; 74:1012-1028. [PMID: 36054915 DOI: 10.1002/iub.2669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Steviol glycosides, the active sweet components of stevia plant, have been recently found to possess a number of therapeutic properties, including some recorded anticancer ones against various cancer cell types (breast, ovarian, cervical, pancreatic, and colon cancer). Our aim was to investigate this anticancer potential on the two most commonly used breast cancer cell lines which differ in the phenotype and estrogen receptor (ER) status: the low metastatic, ERα+ MCF-7 and the highly metastatic, ERα-/ERβ+ MDA-MB-231. Specifically, glycosides' effect was studied on cancer cells': (a) viability, (b) functionality (proliferation, migration, and adhesion), and (c) gene expression (mRNA level) of crucial molecules implicated in cancer's pathophysiology. Results showed that steviol glycosides induced cell death in both cell lines, in the first 24 hr, which was in line with the antiapoptotic BCL2 decrease. However, cells that managed to survive showcased diametrically opposite behavior. The low metastatic ERα+ MCF-7 cells acquired an aggressive phenotype, depicted by the upregulation of all receptors and co-receptors (ESR, PGR, AR, GPER1, EGFR, IGF1R, CD44, SDC2, and SDC4), as well as VIM and MMP14. On the contrary, the highly metastatic ERα-/ERβ+ MDA-MB-231 cells became less aggressive as pointed out by the respective downregulation of EGFR, IGF1R, CD44, and SDC2. Changes observed in gene expression were compatible with altered cell functions. Glycosides increased MCF-7 cells migration and adhesion, but reduced MDA-MB-231 cells migratory and metastatic potential. In conclusion, the above data clearly demonstrate that steviol glycosides have different effects on breast cancer cells according to their ER status, suggesting that steviol glycosides might be examined for their potential anticancer activity against breast cancer, especially triple negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Christos Velesiotis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Department of Chemistry, University of Patras, Patras, Greece
| | - Marinos Kanellakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Department of Chemistry, University of Patras, Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
9
|
Bonfim Neto AP, Cardoso APMM, Silva RDS, Sousa LMMDC, Giometti IC, Binelli M, Bauersachs S, Kowalewski MP, Papa PDC. An approach to uncover the relationship between 17b-estradiol and ESR1/ESR2 ratio in the regulation of canine corpus luteum. Front Vet Sci 2022; 9:885257. [PMID: 35982918 PMCID: PMC9378837 DOI: 10.3389/fvets.2022.885257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022] Open
Abstract
The canine corpus luteum (CL) is able to synthetise, activate and deactivate 17b-estradiol (E2) and also expresses nuclear estrogen receptors in a time-dependent manner during diestrus. Nevertheless, we are still missing a better comprehension of E2 functions in the canine CL, especially regarding the specific roles of estrogen receptor alpha (ERa) and ERb, encoded by ESR1 and 2, respectively. For that purpose, we analyzed transcriptomic data of canine non-pregnant CL collected on days 10, 20, 30, 40, 50 and 60 of diestrus and searched for differentially expressed genes (DEG) containing predicted transcription factor binding sites (TFBS) for ESR1 or ESR2. Based on biological functions of DEG presenting TFBS, expression of select transcripts and corresponding proteins was assessed. Additionally, luteal cells were collected across specific time points during diestrus and specificity of E2 responses was tested using ERa and/or ERb inhibitors. Bioinformatic analyses revealed 517 DEGs containing TFBS, from which 67 for both receptors. In general, abundance of predicted ESR1 targets was greater in the beginning, while abundance of ESR2 targets was greater in the end of diestrus. ESR1/ESR2 ratio shifted from an increasing to a decreasing pattern from day 30 to 40 post ovulation. Specific receptor inhibition suggested an ERa-mediated positive regulation of CL function at the beginning of diestrus and an ERb-mediated effect contributing to luteal regression. In conclusion, our data points toward a broad spectrum of action of E2 and its nuclear receptors, which can also act as transcription factors for other genes regulating canine CL function.
Collapse
Affiliation(s)
| | | | - Renata dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | - Mario Binelli
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Stefan Bauersachs
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Paula de Carvalho Papa
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Paula de Carvalho Papa
| |
Collapse
|
10
|
Liang Y, Besch-Williford C, Hyder SM. The estrogen receptor beta agonist liquiritigenin enhances the inhibitory effects of the cholesterol biosynthesis inhibitor RO 48-8071 on hormone-dependent breast-cancer growth. Breast Cancer Res Treat 2022; 192:53-63. [PMID: 35037188 DOI: 10.1007/s10549-021-06487-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Most hormone-dependent human breast cancers develop resistance to anti-hormone therapy over time. Our goal was to identify novel treatment strategies to avoid this drug resistance and thereby control hormone-dependent breast cancer. METHODS Sulforhodamine B assays were used to measure viability of cultured human breast-cancer cells. BT-474 cell tumor xenografts in nude mice were used to evaluate tumor growth. Immunohistochemistry was used to assess estrogen-receptor and angiogenesis-marker expression, as well as apoptosis, in tumor-xenograft tissues. RESULTS MCF-7 and BT-474 breast-cancer cells treated with either RO 48-8071 <[4'-[6-(Allylmethylamino)hexyloxy]-4-bromo-2'-fluorobenzophenone fumarate] [RO]; a small-molecule inhibitor of oxidosqualene cyclase, a key enzyme in cholesterol biosynthesis> or liquiritigenin [LQ; an estrogen receptor (ER) β agonist] exhibited significantly reduced viability in vitro. RO + LQ treatment further significantly reduced cell viability. Administration of RO, LQ, or RO + LQ significantly inhibited growth of BT-474 tumor xenografts in vivo. RO, LQ, or RO + LQ reduced ERα but induced ER β expression in tumor xenografts. Both compounds significantly reduced angiogenesis-marker expression and increased apoptosis in tumor xenografts; use of RO + LQ significantly enhanced the effects observed with a single agent. CONCLUSION The ERβ ligand LQ significantly enhanced the inhibition of breast-cancer cell viability and tumor-xenograft growth by RO. The anti-tumor properties of RO may in part be due to an off-target effect that reduces ERα and increases ERβ, the latter of which can then interact with LQ to promote anti-proliferative effects. The RO + LQ combination may have value when considering novel treatment strategies for hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Yayun Liang
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65211, USA.,Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | | | - Salman M Hyder
- Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO, 65211, USA. .,Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
11
|
Ceccarelli I, Bioletti L, Peparini S, Solomita E, Ricci C, Casini I, Miceli E, Aloisi AM. Estrogens and phytoestrogens in body functions. Neurosci Biobehav Rev 2021; 132:648-663. [PMID: 34890602 DOI: 10.1016/j.neubiorev.2021.12.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/23/2022]
Abstract
Estrogens are the hormones of reproduction in women as well as of many other important functions in the male and female body. They undergo significant changes in the different phases of life, e.g. during puberty, pregnancy or at menopause/andropause. Phytoestrogens are natural non-steroidal phenolic plant compounds that can mimic the activity of estrogens and their beneficial effects in women and in men. This narrative review summarizes the literature on the physiological role of estrogens and the several potential health benefits of phytoestrogens, with particular attention given to the possible role of phytoestrogens in aging.
Collapse
Affiliation(s)
- Ilaria Ceccarelli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Lucia Bioletti
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Sofia Peparini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Erminia Solomita
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Comasia Ricci
- Department Life Sciences, University of Siena, Siena, Italy
| | - Ilenia Casini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Elisangela Miceli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Anna Maria Aloisi
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
| |
Collapse
|
12
|
Abstract
Estrogen receptors (ERs) are known to play an important role in the proper development of estrogen-sensitive organs, as well as in the development and progression of various types of cancer. ERα, the first ER to be discovered, has been the focus of most cancer research, especially in the context of breast cancer. However, ERβ expression also plays a significant role in cancer pathophysiology, notably its seemingly protective nature and loss of expression with oncogenesis and progression. Although ERβ exhibits antitumor activity in breast, ovarian, and prostate cancer, its expression is associated with disease progression and worse prognosis in lung cancer. The function of ERβ is complicated by the presence of multiple isoforms and single nucleotide polymorphisms, in addition to tissue-specific functions. This mini-review explores current literature on ERβ and its mechanism of action and clinical implications in breast, ovarian, prostate, and lung cancer.
Collapse
Affiliation(s)
- Nicole M Hwang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Research Center, Pittsburgh, PA 15232, USA
| | - Laura P Stabile
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Research Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
13
|
Avellaneda E, Lim A, Moeller S, Marquez J, Escalante Cobb P, Zambrano C, Patel A, Sanchez V, Godde K, Broussard C. HPTE-Induced Embryonic Thymocyte Death and Alteration of Differentiation Is Not Rescued by ERα or GPER Inhibition but Is Exacerbated by Concurrent TCR Signaling. Int J Mol Sci 2021; 22:ijms221810138. [PMID: 34576301 PMCID: PMC8471014 DOI: 10.3390/ijms221810138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Organochlorine pesticides, such as DDT, methoxychlor, and their metabolites, have been characterized as endocrine disrupting chemicals (EDCs); suggesting that their modes of action involve interaction with or abrogation of endogenous endocrine function. This study examined whether embryonic thymocyte death and alteration of differentiation induced by the primary metabolite of methoxychlor, HPTE, rely upon estrogen receptor binding and concurrent T cell receptor signaling. Estrogen receptor inhibition of ERα or GPER did not rescue embryonic thymocyte death induced by HPTE or the model estrogen diethylstilbestrol (DES). Moreover, adverse effects induced by HPTE or DES were worsened by concurrent TCR and CD2 differentiation signaling, compared with EDC exposure post-signaling. Together, these data suggest that HPTE- and DES-induced adverse effects on embryonic thymocytes do not rely solely on ER alpha or GPER but may require both. These results also provide evidence of a potential collaborative signaling mechanism between TCR and estrogen receptors to mediate adverse effects on embryonic thymocytes, as well as highlight a window of sensitivity that modulates EDC exposure severity.
Collapse
Affiliation(s)
- Eddie Avellaneda
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Atalie Lim
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Sara Moeller
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Jacqueline Marquez
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Priscilla Escalante Cobb
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Cristina Zambrano
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA;
| | - Aaditya Patel
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - Victoria Sanchez
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
| | - K. Godde
- Department of Sociology/Anthropology, University of La Verne, La Verne, CA 91750, USA;
| | - Christine Broussard
- Department of Biology, University of La Verne, La Verne, CA 91750, USA; (E.A.); (A.L.); (S.M.); (J.M.); (P.E.C.); (A.P.); (V.S.)
- Correspondence:
| |
Collapse
|
14
|
The Potential Effects of Phytoestrogens: The Role in Neuroprotection. Molecules 2021; 26:molecules26102954. [PMID: 34065647 PMCID: PMC8156305 DOI: 10.3390/molecules26102954] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022] Open
Abstract
Phytoestrogens are naturally occurring non-steroidal phenolic plant compounds. Their structure is similar to 17-β-estradiol, the main female sex hormone. This review offers a concise summary of the current literature on several potential health benefits of phytoestrogens, mainly their neuroprotective effect. Phytoestrogens lower the risk of menopausal symptoms and osteoporosis, as well as cardiovascular disease. They also reduce the risk of brain disease. The effects of phytoestrogens and their derivatives on cancer are mainly due to the inhibition of estrogen synthesis and metabolism, leading to antiangiogenic, antimetastatic, and epigenetic effects. The brain controls the secretion of estrogen (hypothalamus-pituitary-gonads axis). However, it has not been unequivocally established whether estrogen therapy has a neuroprotective effect on brain function. The neuroprotective effects of phytoestrogens seem to be related to both their antioxidant properties and interaction with the estrogen receptor. The possible effects of phytoestrogens on the thyroid cause some concern; nevertheless, generally, no serious side effects have been reported, and these compounds can be recommended as health-promoting food components or supplements.
Collapse
|
15
|
Gómez-Zorita S, González-Arceo M, Fernández-Quintela A, Eseberri I, Trepiana J, Portillo MP. Scientific Evidence Supporting the Beneficial Effects of Isoflavones on Human Health. Nutrients 2020; 12:nu12123853. [PMID: 33348600 PMCID: PMC7766685 DOI: 10.3390/nu12123853] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Isoflavones are phenolic compounds with a chemical structure similar to that of estradiol. They are present in several vegetables, mainly in legumes such as soy, white and red clover, alfalfa and beans. The most significant food source of isoflavones in humans is soy-derived products. Isoflavones could be used as an alternative therapy for pathologies dependent on hormonal disorders such as breast and prostate cancer, cardiovascular diseases, as well as to minimize menopausal symptoms. According to the results gathered in the present review, it can be stated that there is scientific evidence showing the beneficial effect of isoflavones on bone health and thus in the prevention and treatment of osteoporosis on postmenopausal women, although the results do not seem entirely conclusive as there are discrepancies among the studies, probably related to their experimental designs. For this reason, the results should be interpreted with caution, and more randomized clinical trials are required. By contrast, it seems that soy isoflavones do not lead to a meaningful protective effect on cardiovascular risk. Regarding cancer, scientific evidence suggests that isoflavones could be useful in reducing the risk of suffering some types of cancer, such as breast and endometrial cancer, but further studies are needed to confirm these results. Finally, isoflavones could be useful in reducing hot flushes associated with menopause. However, a limitation in this field is that there is still a great heterogeneity among studies. Lastly, with regard to isoflavone consumption safety, it seems that they are safe and that the most common adverse effect is mild and occurs at the gastrointestinal level.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria, Spain
- Bioaraba Health Research Institute, 01002 Vitoria, Spain
| | - Maitane González-Arceo
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria, Spain
- Bioaraba Health Research Institute, 01002 Vitoria, Spain
| | - Itziar Eseberri
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria, Spain
- Bioaraba Health Research Institute, 01002 Vitoria, Spain
- Correspondence: (I.E.); (J.T.)
| | - Jenifer Trepiana
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria, Spain
- Bioaraba Health Research Institute, 01002 Vitoria, Spain
- Correspondence: (I.E.); (J.T.)
| | - María Puy Portillo
- Nutrition and Obesity Group, Department of Pharmacy and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain; (S.G.-Z.); (M.G.-A.); (A.F.-Q.); (M.P.P.)
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, 01006 Vitoria, Spain
- Bioaraba Health Research Institute, 01002 Vitoria, Spain
| |
Collapse
|
16
|
Petrine JCP, Del Bianco-Borges B. The influence of phytoestrogens on different physiological and pathological processes: An overview. Phytother Res 2020; 35:180-197. [PMID: 32780464 DOI: 10.1002/ptr.6816] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/01/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Functional foods have nutritional properties and organic functions, which are beneficial to health. Certain types of functional food components are so-called phytoestrogens, non-steroidal compounds derived from the metabolism of precursors contained in plants, which originate secondary metabotypes known to induce biological responses and by mimicry or modulating the action of endogenous estrogen. These molecules are involved in several physiological and pathological processes related to reproduction, bone remodeling, skin, cardiovascular, nervous, immune systems, and metabolism. This review aimed to present an overview of phytoestrogens regarding their chemical structure, actions, and effects in the organism given several pathologies. Several studies have demonstrated beneficial phytoestrogen actions, such as lipid profile improvement, cognitive function, menopause, oxidative stress, among others. Phytoestrogens effects are not completely elucidated, being necessary future research to understand the exact action mechanisms, whether they are via estrogen receptor or whether other hidden mechanisms produce these effects. Thus, this review makes a general approach to the phytoestrogen actions, beneficial effects, risk and limitations. However, the complexities of biological effects after ingestion of phytoestrogens and the differences in their metabolism and bioavailability indicate that interpretation of either risk or benefits needs to be made with caution.
Collapse
Affiliation(s)
- Jéssica C P Petrine
- Departamento de Ciências da Saúde, Universidade Federal de Lavras, Lavras, Brasil
| | | |
Collapse
|
17
|
Hazarika J, Ganguly M, Borgohain G, Baruah I, Sarma S, Bhuyan P, Mahanta R. Endocrine disruption: molecular interactions of chlorpyrifos and its degradation products with estrogen receptor. Struct Chem 2020. [DOI: 10.1007/s11224-020-01562-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Tang C, Zhang J, Liu P, Zhou Y, Hu Q, Zhong Y, Wang X, Chen L. Chronic exposure to low dose of bisphenol A causes follicular atresia by inhibiting kisspeptin neurons in anteroventral periventricular nucleus in female mice. Neurotoxicology 2020; 79:164-176. [PMID: 32407858 DOI: 10.1016/j.neuro.2020.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 11/16/2022]
Abstract
Bisphenol-A (BPA) is an estrogenic chemical extensively used in industrial and household applications. The present study was conducted to investigate the effect of chronic exposure to BPA on the adult female neuroendocrine system. Herein, we found that expose of adult female mice to BPA (50 μg/kg) by oral gavage for 60 days (BPA mice) prolonged diestrus and decreased serum 17β-estradiol (E2) concentration by reducing the number of antral follicles and corpora luteum. In comparison with controls, the levels of serum luteinizing hormone (LH), follicle stimulating hormone (FSH), hypothalamic gonadotrophin releasing hormone (GnRH) and the expression of kisspeptin in anteroventral periventricular nucleus (AVPV) decreased in BPA mice, which could be reversed by injecting kisspeptin-10 (i.c.v.). Treatment with BPA or estrogen receptor α (ERα) antagonist MPP, but not ERβ antagonist PHTPP inhibited E2-induced AVPV-kisspeptin expression in ovariectomized mice. Use of ERα agonist PPT rather than ERβ agonist DPN enhanced AVPV-kisspepetin expression, which decreased after treatment with BPA. The amplitude of the proestrus LH surge decreased in mice exposed to BPA, but was recovered by administering kisspeptin-10. The present study provides in vivo evidence that chronic exposure to a low dose of BPA suppressed ERα-induced activation of AVPV-kisspeptin neurons, leading to prolonged diestrus and reduced ovulation in adult female mice.
Collapse
Affiliation(s)
- Chuanfeng Tang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jia Zhang
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Peiyu Liu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Yu Zhou
- Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Qiaoyun Hu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Ying Zhong
- Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoli Wang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
19
|
Dunphy KA, Black AL, Roberts AL, Sharma A, Li Z, Suresh S, Browne EP, Arcaro KF, Ser-Dolansky J, Bigelow C, Troester MA, Schneider SS, Makari-Judson G, Crisi GM, Jerry DJ. Inter-Individual Variation in Response to Estrogen in Human Breast Explants. J Mammary Gland Biol Neoplasia 2020; 25:51-68. [PMID: 32152951 PMCID: PMC7147970 DOI: 10.1007/s10911-020-09446-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Exposure to estrogen is strongly associated with increased breast cancer risk. While all women are exposed to estrogen, only 12% are expected to develop breast cancer during their lifetime. These women may be more sensitive to estrogen, as rodent models have demonstrated variability in estrogen sensitivity. Our objective was to determine individual variation in expression of estrogen receptor (ER) and estrogen-induced responses in the normal human breast. Human breast tissue from female donors undergoing reduction mammoplasty surgery were collected for microarray analysis of ER expression. To examine estrogen-induced responses, breast tissue from 23 female donors were cultured ex- vivo in basal or 10 nM 17β-estradiol (E2) media for 4 days. Expression of ER genes (ESR1 and ESR2) increased significantly with age. E2 induced consistent increases in global gene transcription, but expression of target genes AREG, PGR, and TGFβ2 increased significantly only in explants from nulliparous women. E2-treatment did not induce consistent changes in proliferation or radiation induced apoptosis. Responses to estrogen are highly variable among women and not associated with levels of ER expression, suggesting differences in intracellular signaling among individuals. The differences in sensitivity to E2-stimulated responses may contribute to variation in risk of breast cancer.
Collapse
Affiliation(s)
- Karen A Dunphy
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA.
| | - Amye L Black
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Amy L Roberts
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Aman Sharma
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Zida Li
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Sneha Suresh
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Eva P Browne
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Kathleen F Arcaro
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Carol Bigelow
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sallie S Schneider
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Pioneer Valley Life Sciences, Springfield, MA, USA
| | - Grace Makari-Judson
- Division of Hematology-Oncology, University of Massachusetts Medical School/Baystate, Springfield, MA, USA
| | - Giovanna M Crisi
- Department of Pathology, University of Massachusetts Medical School/Baystate, Springfield, MA, USA
| | - D Joseph Jerry
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Pioneer Valley Life Sciences, Springfield, MA, USA
| |
Collapse
|
20
|
Adam AHB, de Haan LHJ, Estruch IM, Hooiveld GJEJ, Louisse J, Rietjens IMCM. Estrogen receptor alpha (ERα)-mediated coregulator binding and gene expression discriminates the toxic ERα agonist diethylstilbestrol (DES) from the endogenous ERα agonist 17β-estradiol (E2). Cell Biol Toxicol 2020; 36:417-435. [PMID: 32088792 PMCID: PMC7505815 DOI: 10.1007/s10565-020-09516-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/28/2020] [Indexed: 12/29/2022]
Abstract
Diethylstilbestrol (DES) is a synthetic estrogen and proven human teratogen and carcinogen reported to act via the estrogen receptor α (ERα). Since the endogenous ERα ligand 17β-estradiol (E2) does not show these adverse effects to a similar extent, we hypothesized that DES' interaction with the ERα differs from that of E2. The current study aimed to investigate possible differences between DES and E2 using in vitro assays that detect ERα-mediated effects, including ERα-mediated reporter gene expression, ERα-mediated breast cancer cell (T47D) proliferation and ERα-coregulator interactions and gene expression in T47D cells. Results obtained indicate that DES and E2 activate ERα-mediated reporter gene transcription and T47D cell proliferation in a similar way. However, significant differences between DES- and E2-induced binding of the ERα to 15 coregulator motifs and in transcriptomic signatures obtained in the T47D cells were observed. It is concluded that differences observed in binding of the ERα with several co-repressor motifs, in downregulation of genes involved in histone deacetylation and DNA methylation and in upregulation of CYP26A1 and CYP26B1 contribute to the differential effects reported for DES and E2.
Collapse
Affiliation(s)
- Aziza Hussein Bakheit Adam
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands.
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Ignacio Miro Estruch
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Guido J E J Hooiveld
- Division of Human Nutrition and Health, Wageningen University and Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Jochem Louisse
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| |
Collapse
|
21
|
Factors affecting the fate of the canine corpus luteum: Potential contributors to pregnancy and non-pregnancy. Theriogenology 2020; 150:339-346. [PMID: 32089321 DOI: 10.1016/j.theriogenology.2020.01.081] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022]
Abstract
The fate of the canine corpus luteum (CL) differs from that of other domestic species: beyond the extended luteal regression observed in both pregnant and non-pregnant cycles, active luteolysis is observed only in pregnant dogs. Luteal regression in the absence of pregnancy lacks a luteolytic trigger. The CL lifespan during pregnancy is around 60 days, as long as that of the cyclic CL. Although they are already available in the first half of diestrus, LH and especially prolactin (PRL) play a decisive luteotropic role from approximately day 25 post-ovulation onwards. Nevertheless, many locally-produced factors are orchestrated to ensure a fully functional CL, which in the bitch produces progesterone (P4), 17b-estradiol, and other local regulators. Recently, insulin has been described as another luteotropic factor in this species, able to increase glucose uptake in luteal cells and contribute to steroid biosynthesis. The locally-produced PGE2 is also a potent luteotropic factor in the first half of diestrus, promoting STAR expression, as are also proliferating, vasoactive- and immunomodulatory factors. These, in turn, all contribute to the formation and maintenance of the canine CL. Meanwhile PGF2a, produced by the utero-placental compartment, participates actively in triggering pre-partum luteolysis. Cytokines play different roles, either contributing as luteotropic or as acute inflammation molecules. So far, the one clinically most efficient mechanism of interrupting a pregnancy in the dog is to block P4 receptors, using an antigestagen (e.g., aglepristone) in the second half of diestrus. To enhance the chances of pregnancy, however, several luteotropic factors could be used.
Collapse
|
22
|
Toporova L, Balaguer P. Nuclear receptors are the major targets of endocrine disrupting chemicals. Mol Cell Endocrinol 2020; 502:110665. [PMID: 31760044 DOI: 10.1016/j.mce.2019.110665] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
Abstract
Endocrine disrupting chemicals (EDCs) are exogenous substances that are suspected to cause adverse effects in the endocrine system mainly by acting through their interaction with nuclear receptors such as the estrogen receptors α and β (ERα and ERβ), the androgen receptor (AR), the pregnan X receptor (PXR), the peroxisome proliferator activated receptors α and γ (PPARα, PPARγ) and the thyroid receptors α and β (TRα and TRβ). More recently, the retinoid X receptors (RXRα, RXRβ and RXRγ), the constitutive androstane receptor (CAR) and the estrogen related receptor γ (ERRγ) have also been identified as targets of EDCs. Finally, nuclear receptors still poorly studied for their interaction with environmental ligands such as the progesterone receptor (PR), the mineralocorticoid receptor (MR), the glucocorticoid receptor (GR), the retinoic acid receptors (RAR α, RARβ and RARγ), the farnesoid X receptor (FXR) and the liver X receptors α and β (LXRα and LXβ) as well are suspected targets of EDCs. Humans are generally exposed to low doses of pollutants, therefore the aim of current research is to identify the targets of EDCs at environmental concentrations. In this review, we analyze recent works referring that nuclear receptors are targets of EDCs and we highlight which EDCs are able to act at low concentrations.
Collapse
Affiliation(s)
- Lucia Toporova
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| |
Collapse
|
23
|
A nutraceutical composition containing diosmin and hesperidin has osteogenic and anti-resorptive effects and expands the anabolic window of teriparatide. Biomed Pharmacother 2019; 118:109207. [DOI: 10.1016/j.biopha.2019.109207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/27/2019] [Accepted: 07/02/2019] [Indexed: 02/05/2023] Open
|
24
|
Whitman NA, Lin ZW, Kenney RM, Albertini L, Lockett MR. Hypoxia differentially regulates estrogen receptor alpha in 2D and 3D culture formats. Arch Biochem Biophys 2019; 671:8-17. [PMID: 31163125 PMCID: PMC6688900 DOI: 10.1016/j.abb.2019.05.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/24/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Hypoxia is a common feature in solid tumors. Clinical samples show a positive correlation between the expression of the hypoxia-inducible factor HIF-1α and estrogen receptor alpha (ERα) and a negative correlation between HIF-1α and hormone sensitivity. Results from monolayer cultures are in contention with clinical observations, showing that ER (+) cell lines no longer express ERα under hypoxic conditions (1% O2). Here, we compared the impact of hypoxia on the ERα signaling pathway for T47D cells in a 2D and 3D culture format. In the 2D format, the cells were cultured as monolayers. In the 3D format, paper-based scaffolds supported cells suspended in a collagen matrix. Using ELISA, Western blot, and immunofluorescence measurements, we show that hypoxia differentially regulates ERα protein levels in a culture environment-dependent manner. In the 2D format, the protein levels are significantly decreased in hypoxia. In the 3D format, the protein levels are maintained in hypoxia. Hypoxia reduced ERα transcriptional activation in both culture formats. These results highlight the importance of considering tissue dimensionality for in vitro studies. They also show that ERα protein levels in hypoxia are not an accurate indicator of ERα transcriptional activity, and confirm that a positive stain for ERα in a clinical sample may not necessarily indicate hormone sensitivity.
Collapse
Affiliation(s)
- Nathan A Whitman
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Zhi-Wei Lin
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Rachael M Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Leonardo Albertini
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Matthew R Lockett
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC, 27599-7295, USA.
| |
Collapse
|
25
|
Eze UA, Huntriss J, Routledge MN, Gong YY, Connolly L. The effect of individual and mixtures of mycotoxins and persistent organochloride pesticides on oestrogen receptor transcriptional activation using in vitro reporter gene assays. Food Chem Toxicol 2019; 130:68-78. [PMID: 31082460 DOI: 10.1016/j.fct.2019.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
The mycotoxins zearalenone (ZEN) and alpha-zearalenone (α-ZOL), which are common contaminants of agri-food products, are known for their oestrogenic potential. In addition to mycotoxins, food may also contain pesticides with oestrogenic properties such as 1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane (p,p'-DDT) and 1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene (p,p'-DDE), raising the question on the potential effects of individual and combinations of these xeno-oestrogens on the action of natural oestrogens. The present study employed a mammalian reporter gene assay to assess the effects individual and binary combinations of these environmental and food-borne contaminants on oestrogen nuclear receptor (ER) transactivation. As expected, α-ZOL and ZEN exhibited the strongest oestrogenic potency (EC50: 0.27 ± 0.121 nM and 1.32 ± 0.0956 nM, respectively) whereas p,p'-DDT and p,p'-DDE had weak ER agonistic activity with the maximal response of 28.70 ± 2.97% and 18.65 ± 1.77%, respectively. Concurrent treatment of the mycotoxins and/or pesticides, individually or in binary combination, with 17β-oestradiol (E2) showed either additive, synergistic or antagonistic interactive effects on E2-mediated ER response, depending on the combination ratios, the concentration range of xeno-oestrogens, and the concentration of E2. This study highlights the importance of assessing the mixture effects of chemical contaminants in risk assessment, especially in the area of reproductive and developmental toxicity.
Collapse
Affiliation(s)
- Ukpai A Eze
- School of Food Science and Nutrition, Food Science Building, University of Leeds, LS2 9JT, UK; Department of Medical Laboratory Sciences, Faculty of Health Sciences, Ebonyi State University, P. M. B. 053, Abakaliki, Nigeria
| | - John Huntriss
- Division of Reproduction and Early Development, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, LS2 9JT, UK
| | - Michael N Routledge
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, LS2 9JT, UK
| | - Yun Yun Gong
- School of Food Science and Nutrition, Food Science Building, University of Leeds, LS2 9JT, UK; Department of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Ministry of Health, Beijing, 100021, PR China.
| | - Lisa Connolly
- Institute for Global Food Security, School of Biological Sciences, Queens University Belfast, Northern Ireland, BT9 5AF, UK
| |
Collapse
|
26
|
Křížová L, Dadáková K, Kašparovská J, Kašparovský T. Isoflavones. Molecules 2019; 24:E1076. [PMID: 30893792 PMCID: PMC6470817 DOI: 10.3390/molecules24061076] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Phytoestrogens are naturally occurring nonsteroidal phenolic plant compounds that, due to their molecular structure and size, resemble vertebrate steroids estrogens. This review is focused on plant flavonoids isoflavones, which are ranked among the most estrogenic compounds. The main dietary sources of isoflavones for humans are soybean and soybean products, which contain mainly daidzein and genistein. When they are consumed, they exert estrogenic and/or antiestrogenic effects. Isoflavones are considered chemoprotective and can be used as an alternative therapy for a wide range of hormonal disorders, including several cancer types, namely breast cancer and prostate cancer, cardiovascular diseases, osteoporosis, or menopausal symptoms. On the other hand, isoflavones may also be considered endocrine disruptors with possible negative influences on the state of health in a certain part of the population or on the environment. This review deals with isoflavone classification, structure, and occurrence, with their metabolism, biological, and health effects in humans and animals, and with their utilization and potential risks.
Collapse
Affiliation(s)
- Ludmila Křížová
- Department of Biochemistry, Faculty of Science, Masaryk University, Kotlarska 2, 61137 Brno, Czech Republic.
| | - Kateřina Dadáková
- Department of Biochemistry, Faculty of Science, Masaryk University, Kotlarska 2, 61137 Brno, Czech Republic.
| | - Jitka Kašparovská
- Department of Biochemistry, Faculty of Science, Masaryk University, Kotlarska 2, 61137 Brno, Czech Republic.
| | - Tomáš Kašparovský
- Department of Biochemistry, Faculty of Science, Masaryk University, Kotlarska 2, 61137 Brno, Czech Republic.
| |
Collapse
|
27
|
Tarnow P, Tralau T, Luch A. Chemical activation of estrogen and aryl hydrocarbon receptor signaling pathways and their interaction in toxicology and metabolism. Expert Opin Drug Metab Toxicol 2019; 15:219-229. [PMID: 30644759 DOI: 10.1080/17425255.2019.1569627] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Estrogen receptors (ERs) and the arylhydrocarbon receptor (AHR) are ligand-activated transcription factors that regulate the expression of genes involved in many physiological processes. With both receptors binding a broad range of natural and anthropogenic ligands, they are molecular targets for many substances, raising concerns for possible health effects. Areas covered: This review shall give a brief overview on the physiological functions of both receptors including their underlying molecular mechanisms. It summarizes the interaction of the respective signaling pathways including impacts on metabolism of endogenous estrogens, transcriptional interference, inhibitory crosstalk, and proteasomal degradation. Also addressed are the AHR dependent formation of estrogenic metabolites from polycyclic aromatic hydrocarbons and the possible impact of the ER/AHR crosstalk in the context of drug metabolism. Expert opinion: Despite decade-long research, the physiological role of the AHR and ER as well as the implications of their complex mutual crosstalk remain to be determined as do resulting potential impacts on human health. With more and more endogenous AHR ligands being discovered, future research should hence systematically address the potential impact of such substances on estrogen signaling. The intimate link between these two pathways and the genes regulated therein bears the potential for impacts on drug metabolism and human health.
Collapse
Affiliation(s)
- Patrick Tarnow
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| | - Tewes Tralau
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| | - Andreas Luch
- a Department of Chemical & Product Safety , German Federal Institute for Risk Assessment (BfR) , Berlin , Germany
| |
Collapse
|
28
|
Park YJ, Zheng H, Kwak JH, Chung KH. Sesquiterpenes from Cyperus rotundus and 4α,5α-oxidoeudesm-11-en-3-one as a potential selective estrogen receptor modulator. Biomed Pharmacother 2018; 109:1313-1318. [PMID: 30551381 DOI: 10.1016/j.biopha.2018.10.186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/10/2023] Open
Abstract
Estrogenic activity-oriented fractionation and purification of methanol extract from the rhizome of Cyperus rotundus, a well-known traditional herbal medicine, led to the isolation of six sesquiterpenes. 4α,5α-Oxidoeudesm-11-en-3-one (2) and cyper-11-ene-3,4-dione (3) together with four known sesquiterpenes, cyperotundone (1), caryophyllene α-oxide (4), α-cyperone (5), and isocyperol (6) were obtained from the hexane and dichloromethane fractions. Compounds 2 and 3 were newly isolated from natural resources in particular. To identify the possible use of isolated compounds as an alternative to hormone replacement therapy (HRT), estrogenic activity was evaluated by E-screen assay on MCF-7 BUS cells. Among the all isolated compounds from the rhizome of Cyperus rotundus, newly isolated from natural resource, 2 exhibited the most potent estrogenic activity. In an estrogen sensitive reporter gene assay, 2 significantly increased transcriptional activities. As a phytoestrogen, 2 was assessed by investigating dual action on ER-α and ER-β in competitive binding assay. It was found that 2 exerted higher binding affinity to ER-β than ER-α and it also showed both estrogenic and antiestrogenic effects depending on the E2 concentration. Our results indicate that newly isolated from Cyperus rotundus, 2 has biphasic activities on estrogen receptors which could be useful as an alternative HRT.
Collapse
Affiliation(s)
- Yong Joo Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hailing Zheng
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Kyu Hyuck Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
29
|
Islam MA, Hooiveld GJEJ, van den Berg JHJ, van der Velpen V, Murk AJ, Rietjens IMCM, van Leeuwen FXR. Soy supplementation: Impact on gene expression in different tissues of ovariectomized rats and evaluation of the rat model to predict (post)menopausal health effect. Toxicol Rep 2018; 5:1087-1097. [PMID: 30425930 PMCID: PMC6222031 DOI: 10.1016/j.toxrep.2018.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 09/29/2018] [Accepted: 10/18/2018] [Indexed: 11/06/2022] Open
Abstract
The usefulness of PBMC gene expressions as a surrogate tissue for risk assessment is questionable. SIF in a dose of 2 mg/kg b.w/day is not able to influence ERGs in target tissues such as breast and uterus. Plasma concentrations of SIF after 8 weeks oral exposure similar as the recommended dose for humans do not proliferate cells in in vitro cellular models. The ovariectomized rat is probably not a good model to predict human risk or benefit assessment of SIF in human.
This toxicogenomic study was conducted to predict (post)menopausal human health effects of commercial soy supplementation using ovariectomized rats as a model. Different target tissues (i.e. breast, uterus and sternum) and non-target tissues (i.e. peripheral blood mononuclear cells (PBMC), adipose and liver) of ovariectomized F344 rats exposed to a commercially available soy supplement for eight weeks, were investigated. Changes in gene expression in these tissues were analysed using whole-genome microarray analysis. No correlation in changes in gene expression were observed among different tissues, indicating tissue specific effects of soy isoflavone supplementation. Out of 87 well-established estrogen responsive genes (ERGs), only 19 were found to be significantly regulated (p < 0.05) in different tissues, particularly in liver, adipose and uterus tissues. Surprisingly, no ERGs were significantly regulated in estrogen sensitive breast and sternum tissues. The changes in gene expression in PBMC and adipose tissue in rats were compared with those in (post)menopausal female volunteers who received the same supplement in a similar oral dose and exposure duration in human intervention studies. No correlation in changes in gene expression between rats and humans was observed. Although receiving a similar dose, in humans the plasma levels expressed as total free aglycones were several folds higher than in the rat. Therefore, the overall results in young ovariectomized female F344 rats indicated that using rat transcriptomic data does not provide a suitable model for human risk or benefit analysis of soy isoflavone supplementation.
Collapse
Affiliation(s)
- Mohammed A Islam
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands.,Department of Agricultural Chemistry, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Guido J E J Hooiveld
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HE Wageningen, the Netherlands
| | | | - Vera van der Velpen
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HE Wageningen, the Netherlands.,Metabolomics Service and Research Unit, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Albertinka J Murk
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands.,Sub-department of Environmental Technology, Wageningen University, the Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - F X Rolaf van Leeuwen
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| |
Collapse
|
30
|
Song W, Liu QS, Sun Z, Yang X, Zhou Q, Jiang G. Polyfluorinated iodine alkanes regulated distinct breast cancer cell progression through binding with estrogen receptor alpha or beta isoforms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 239:300-307. [PMID: 29665550 DOI: 10.1016/j.envpol.2018.04.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
Polyfluorinated iodine alkanes (PFIs) are a kind of emerging chemicals with endocrine disrupting effects. Based on the different binding preferences of PFIs to estrogen receptor alpha and beta isoforms (ERα and β), two representative PFIs, dodecafluoro-1,6-diiodohexane (PFHxDI) and tridecafluorohexyl iodide (PFHxI), were selected to evaluate their effects on the proliferation of two kinds of breast cancer cells with different ERα/β expression levels, MCF-7 and T47D. The cell viability assay showed PFHxDI could cause higher cellular toxicity than did PFHxI in both MCF-7 and T47D. MCF-7 with relatively higher ERα/β expression ratio was more vulnerable to the cytotoxic treatments of PFHxI and PFHxDI when compared with T47D cells with relatively lower ERα/β expression ratio. EdU incorporation and cell cycle analysis revealed that, similar to 17β-estrodiol (E2), non-cytotoxic levels of PFHxDI could significantly promote the proliferation of MCF-7 by increasing cell population at S phase (p < 0.01), while T47D proliferation was not influenced by PFHxI exposure due to cell cycle arrest at G2/M phase. The cellular responses caused by estrogenic PFIs were dominantly mediated by their preferential binding affinities for ER isoforms, which would be helpful in the accurate assessment for their potential influences on the breast cancer progression.
Collapse
Affiliation(s)
- Wenting Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; Medical College, Henan Polytechnic University, Jiaozuo, 454000, PR China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
31
|
Cook MT. Mechanism of metastasis suppression by luteolin in breast cancer. BREAST CANCER-TARGETS AND THERAPY 2018; 10:89-100. [PMID: 29928143 PMCID: PMC6003288 DOI: 10.2147/bctt.s144202] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metastatic breast cancer is typically an extremely aggressive cancer with poor prognosis. Metastasis requires the orchestration of homeostatic factors and cellular programs, many of which are potential therapeutic targets. Luteolin (2-[3,4-dihydroxyphenyl]-5,7-dihydroxy-4-chromenone), is a naturally occurring flavonoid found in fruits and vegetables that exhibits many anticancer properties. Luteolin obstructs metastasis through both direct and indirect mechanisms. For instance, luteolin may suppress breast cancer invasion by acting as an antiangiogenic therapeutic inhibiting VEGF production and its receptor’s activity. Furthermore, luteolin decreases epithelial–mesenchymal transition markers and metastatic proclivity. Luteolin also acts as an antiproliferative by suppressing receptor tyrosine-kinase activity and apoptosis, both of which could prevent incipient colonization of breast cancer. Many of these antimetastatic characteristics accredited to luteolin are likely functionally related. For instance, the PI3K/Akt pathway, which is impeded by luteolin, has several downstream programs involved in increased proliferation, survival, and metastatic potential in breast cancer. In this review, luteolin’s ability to ameliorate breast cancer is summarized. The paper also offers insight into the molecular mechanisms by which luteolin may suppress breast cancer metastasis.
Collapse
Affiliation(s)
- Matthew T Cook
- Department of Biology, Washburn University, Topeka, KS, USA
| |
Collapse
|
32
|
LaPlante CD, Bansal R, Dunphy KA, Jerry DJ, Vandenberg LN. Oxybenzone Alters Mammary Gland Morphology in Mice Exposed During Pregnancy and Lactation. J Endocr Soc 2018; 2:903-921. [PMID: 30057971 PMCID: PMC6057512 DOI: 10.1210/js.2018-00024] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/08/2018] [Indexed: 02/08/2023] Open
Abstract
Hormones and endocrine-disrupting chemicals are generally thought to have permanent “organizational” effects when exposures occur during development but not adulthood. Yet, an increasing number of studies have shown that pregnant females are disrupted by endocrine-disrupting chemical exposures, with some effects that are permanent. Here, we examined the long-term effects of exposure to oxybenzone, an estrogenic chemical found in sunscreen and personal care products, on the morphology of the mammary gland in mice exposed during pregnancy and lactation. Female mice were exposed to vehicle or 30, 212, or 3000 µg oxybenzone/kg/d, from pregnancy day 0 until weaning. A nulliparous group, receiving vehicle treatment, was also evaluated. Mammary glands were collected 5 weeks after involution for whole-mount, histological, immunohistochemical, and molecular analyses. Exposure to 3000 µg oxybenzone/kg/d induced permanent changes to ductal density that was significantly different from both the nulliparous and vehicle groups. The two highest doses of oxybenzone similarly induced an intermediate phenotype for expression of progesterone receptor. A monotonic, dose-dependent increase in cell proliferation was also observed in the oxybenzone-treated females, becoming statistically significant at the highest dose. Finally, oxybenzone exposure induced an intermediate phenotype for Esr1 expression in all oxybenzone-treated groups. These data suggest that oxybenzone, at doses relevant to human exposures, produces long-lasting alterations to mammary gland morphology and function. Further studies are needed to determine if exposure to this chemical during pregnancy and lactation will interfere with the known protection that pregnancy provides against breast cancer.
Collapse
Affiliation(s)
- Charlotte D LaPlante
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Ruby Bansal
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Karen A Dunphy
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - D Joseph Jerry
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| |
Collapse
|
33
|
Liu JT, Do TJ, Simmons CJ, Lynch JC, Gu W, Ma ZX, Xu W, Tang W. Total synthesis of diptoindonesin G and its analogues as selective modulators of estrogen receptors. Org Biomol Chem 2018; 14:8927-8930. [PMID: 27714255 DOI: 10.1039/c6ob01657j] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have developed a versatile synthetic strategy for the synthesis of the natural product diptoindonesin G and its analogues as selective modulators of estrogen receptors. The strategy involves a regioselective dehydrative cyclization of arylacetals, a regioselective bromination of benzofurans, a sequential cross-coupling of bromo-benzofurans with aryl boronic acids, and a BBr3-mediated tandem cyclization and demethylation. Preliminary biological studies uncovered the critical and dispensable phenolic hydroxyl groups in the natural product and also revealed unexpected selectivity for isoforms of estrogen receptor.
Collapse
Affiliation(s)
- Ji-Tian Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA and School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Truman J Do
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Christopher J Simmons
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA and Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John C Lynch
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wen Gu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA and Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Zhi-Xiong Ma
- Medicinal Chemistry Center, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Wei Xu
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA and Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA and Medicinal Chemistry Center, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
34
|
Song W, Zhao L, Sun Z, Yang X, Zhou Q, Jiang G. A novel high throughput screening assay for binding affinities of perfluoroalkyl iodide for estrogen receptor alpha and beta isoforms. Talanta 2017; 175:413-420. [PMID: 28842010 DOI: 10.1016/j.talanta.2017.07.068] [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: 05/31/2017] [Revised: 07/17/2017] [Accepted: 07/23/2017] [Indexed: 11/26/2022]
Abstract
Contaminants of emerging concern are continuously increasing, which makes it important to develop high throughput screening techniques for the evaluation of their potential biological effects, especially endocrine disrupting effects, which would directly influence the population dynamics in environment. A novel competitive binding assay based on enzyme fragmentation complementation technology was established to screen the binding affinities of emerging chemicals for estrogen receptor (ER) α or β isoforms. Exogenous compounds could compete with the fragment (ED-ES) of genetically engineered β-galactosidase enzyme (β-gal) for the binding to ERα or β, thus quantitatively altering the formation of enzymatically active β-gal and the hydrolysis of luminescent substrate. According to the monitoring of luminescence curves and the optimization of ERα or β concentrations, it was found that luminescent signals were sustainably emitted for 9h, and 40nM ERα or β in the system would lead to the most sensitive luminescence response. Using 17β-estrodiol (E2) and genistein as the representative estrogenic hormones, their binding affinities for ERα and β were evaluated. The results were consistent with those determined by traditional methods, which confirmed the reliability of this competitive binding assay based on β-gal. Four polyfluorinated iodine alkanes (PFIs) with specific structural characteristics in iodine substitution and carbon chain length were screened, and the results showed diverse binding affinities and different preferences of these chemicals to ERα or β isoforms. The binding affinities of PFIs for ERα were consistent with the result from MVLN transcriptional reporter assay. Overall, the competitive binding assay presented in this study provided a promising alternative to high throughput screening of emerging chemicals with estrogenic effects, which would be important in explanation of their potential toxicological effects and human exposure risks.
Collapse
Affiliation(s)
- Wenting Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; Medical College, Henan Polytechnic University, Jiaozuo 454000, PR China
| | - Lixia Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zhendong Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
35
|
Rietjens IMCM, Louisse J, Beekmann K. The potential health effects of dietary phytoestrogens. Br J Pharmacol 2017; 174:1263-1280. [PMID: 27723080 PMCID: PMC5429336 DOI: 10.1111/bph.13622] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/04/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022] Open
Abstract
Phytoestrogens are plant-derived dietary compounds with structural similarity to 17-β-oestradiol (E2), the primary female sex hormone. This structural similarity to E2 enables phytoestrogens to cause (anti)oestrogenic effects by binding to the oestrogen receptors. The aim of the present review is to present a state-of-the-art overview of the potential health effects of dietary phytoestrogens. Various beneficial health effects have been ascribed to phytoestrogens, such as a lowered risk of menopausal symptoms like hot flushes and osteoporosis, lowered risks of cardiovascular disease, obesity, metabolic syndrome and type 2 diabetes, brain function disorders, breast cancer, prostate cancer, bowel cancer and other cancers. In contrast to these beneficial health claims, the (anti)oestrogenic properties of phytoestrogens have also raised concerns since they might act as endocrine disruptors, indicating a potential to cause adverse health effects. The literature overview presented in this paper illustrates that several potential health benefits of phytoestrogens have been reported but that, given the data on potential adverse health effects, the current evidence on these beneficial health effects is not so obvious that they clearly outweigh the possible health risks. Furthermore, the data currently available are not sufficient to support a more refined (semi) quantitative risk-benefit analysis. This implies that a definite conclusion on possible beneficial health effects of phytoestrogens cannot be made. LINKED ARTICLES This article is part of a themed section on Principles of Pharmacological Research of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.11/issuetoc.
Collapse
Affiliation(s)
| | - Jochem Louisse
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| | - Karsten Beekmann
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| |
Collapse
|
36
|
Liu Y, Hou Y, Ma L, Sun C, Pan J, Yang Y, Zhou H, Zhang J. Regulation of semaphorin 4D expression and cell proliferation of ovarian cancer by ERalpha and ERbeta. ACTA ACUST UNITED AC 2017; 50:e6057. [PMID: 28225892 PMCID: PMC5333722 DOI: 10.1590/1414-431x20166057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/13/2016] [Indexed: 12/01/2022]
Abstract
Ovarian cancer is one of the most common malignancies in women. Semaphorin 4D (sema 4D) is involved in the progress of multiple cancers. In the presence of estrogen-like ligands, estrogen receptors (ERα and ERβ) participate in the progress of breast and ovarian cancers by transcriptional regulation. The aim of the study was to investigate the role of sema 4D and elucidate the regulatory pattern of ERα and ERβ on sema 4D expression in ovarian cancers. Sema 4D levels were up-regulated in ovarian cancer SKOV-3 cells. Patients with malignant ovarian cancers had significantly higher sema 4D levels than controls, suggesting an oncogene role of sema 4D in ovarian cancer. ERα expressions were up-regulated in SKOV-3 cells compared with normal ovarian IOSE80 epithelial cells. Conversely, down-regulation of ERβ was observed in SKOV-3 cells. Forced over-expression of ERα and ERβ in SKOV-3 cells was manipulated to establish ERα+ and ERβ+ SKOV-3 cell lines. Incubation of ERα+ SKOV-3 cells with ERs agonist 17β-estradiol (E2) significantly enhanced sema 4D expression and rate of cell proliferation. Incubated with E2, ERβ+ SKOV-3 cells showed lower sema 4D expression and cell proliferation. Blocking ERα and ERβ activities with ICI182-780 inhibitor, sema 4D expressions and cell proliferation of ERα+ and ERβ+ SKOV-3 cells were recovered to control levels. Taken together, the data showed that sema 4D expression was positively correlated with the progress of ovarian cancer. ERα positively regulated sema 4D expression and accelerated cell proliferation. ERβ negatively regulated sema 4D expression and inhibited cell multiplication.
Collapse
Affiliation(s)
- Y Liu
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Hou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - L Ma
- Department of Reproduction and Genetics, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - C Sun
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Pan
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Yang
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - H Zhou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Zhang
- Department of General Surgery, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
37
|
Wolff M, Kosyna FK, Dunst J, Jelkmann W, Depping R. Impact of hypoxia inducible factors on estrogen receptor expression in breast cancer cells. Arch Biochem Biophys 2016; 613:23-30. [PMID: 27823907 DOI: 10.1016/j.abb.2016.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/31/2016] [Accepted: 11/03/2016] [Indexed: 12/29/2022]
Abstract
In women breast cancer is still the most commonly diagnosed cancer. This type of cancer is classified as a hormone-dependent tumor. Estrogen receptor (ER) expression and functional status contribute to breast cancer development and progression. Another important factor associated with cancer is hypoxia which is defined as the state of reduced oxygen availability in tissues. Intratumoral hypoxia results in the activation of the hypoxia inducible factors (HIFs). HIFs are heterodimeric transcription factors involved in the regulation of many cellular processes, such as angiogenesis, anaerobic metabolism, cell proliferation/survival, and promotion of metastasis. In this study we evaluated the interplay between hypoxia, HIF stabilization and the ER-α/β-ratio in several ER-positive breast cancer cell lines. Hypoxia was shown to inhibit ER expression in ER-positive breast cancer cells. Further experiments using the hypoxia mimetic CoCl2 and HIF-1α knockdown cells indicated that the influence of hypoxia on breast cancer cells involves other pathways than the molecular oxygen sensing pathway. Moreover, we demonstrated that MCF-7 cells in long-term culture lost part of their ability to respond to hypoxic incubation. Understanding the relationships between HIF, ER-α and ER-β expression holds the promise of the development of new therapeutic agents and may provide future advances in prognosis.
Collapse
Affiliation(s)
- Matthias Wolff
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany; Klinik für Strahlentherapie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Germany
| | | | - Jürgen Dunst
- Klinik für Strahlentherapie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Germany
| | - Wolfgang Jelkmann
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany
| | - Reinhard Depping
- Institute of Physiology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Germany.
| |
Collapse
|
38
|
Mo XM, Li L, Zhu P, Dai YJ, Zhao TT, Liao LY, Chen GG, Liu ZM. Up-regulation of Hsp27 by ERα/Sp1 facilitates proliferation and confers resistance to apoptosis in human papillary thyroid cancer cells. Mol Cell Endocrinol 2016; 431:71-87. [PMID: 27179757 DOI: 10.1016/j.mce.2016.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 04/15/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023]
Abstract
17β-estradiol (E2) has been suggested to play a role in the development and progression of papillary thyroid cancer. Heat shock protein 27 (Hsp27) is a member of the Hsp family that is responsible for cell survival under stressful conditions. Previous studies have shown that the 5'-promoter region of Hsp27 gene contains a specificity protein-1 (Spl) and estrogen response element half-site (ERE-half), which contributes to Hsp27 induction by E2 in breast cancer cells. However, it is unclear whether Hsp27 can be up-regulated by E2 and which estrogen receptor (ER) isoform and tethered transcription factor are involved in this regulation in papillary thyroid cancer cells. In the present study, we demonstrated that Hsp27 can be effectively up-regulated by E2 at mRNA and protein levels in human K1 and BCPAP papillary thyroid cancer cells which have more than two times higher level of ERα than that of ERβ. The up-regulation of Hsp27 by E2 is mediated by ERα/Sp1 and ERβ has repressive effect on this ERα/Sp1-mediated up-regulation of Hsp27. Moreover, we showed that the up-regulation of Hsp27 by ERα/Sp1 facilitates proliferation and confers resistance to apoptosis through interaction with procaspase-3. Targeting this pathway may be a potential strategy for therapy of papillary thyroid cancer.
Collapse
Affiliation(s)
- Xiao-Mei Mo
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Li Li
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Yu-Jie Dai
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ting-Ting Zhao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ling-Yao Liao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China
| | - Zhi-Min Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
39
|
Maltais R, Trottier A, Barbeau X, Lagüe P, Perreault M, Thériault JF, Lin SX, Poirier D. Impact of structural modifications at positions 13, 16 and 17 of 16β-(m-carbamoylbenzyl)-estradiol on 17β-hydroxysteroid dehydrogenase type 1 inhibition and estrogenic activity. J Steroid Biochem Mol Biol 2016; 161:24-35. [PMID: 26519987 DOI: 10.1016/j.jsbmb.2015.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 09/11/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
Abstract
The chemical synthesis of four stereoisomers (compounds 5a-d) of 16β-(m-carbamoylbenzyl)-estradiol, a potent reversible inhibitor of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1), and two intermediates (compounds 3a and b) was performed. Assignment of all nuclear magnetic resonance signals confirmed the stereochemistry at positions 13, 16 and 17. Nuclear overhauser effects showed clear correlations supporting a C-ring chair conformation for 5a and b and a C-ring boat conformation for 5c and d. These compounds were tested as 17β-HSD1 inhibitors and to assess their proliferative activity on estrogen-sensitive breast cancer cells (T-47D) and androgen-sensitive prostate cancer cells (LAPC-4). Steroid derivative 5a showed the best inhibitory activity for the transformation of estrone to estradiol (95, 82 and 27%, at 10, 1 and 0.1μM, respectively), but like the other isomers 5c and d, it was found to be estrogenic. The intermediate 3a, however, was weakly estrogenic at 1μM, not at all at 0.1μM, and showed an interesting inhibitory potency on 17β-HSD1 (90, 59 and 22%, at 10, 1 and 0.1μM, respectively). As expected, no compound showed an androgenic activity. The binding modes for compounds 3a and b, 5a-d and CC-156 were evaluated from molecular modeling. While the non-polar interactions were conserved for all the inhibitors in their binding to 17β-HSD1, differences in polar interactions and in binding conformational energies correlated to the inhibitory potencies.
Collapse
Affiliation(s)
- René Maltais
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Alexandre Trottier
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Xavier Barbeau
- Département de chimie, Institut de biologie intégrative et des systèmes (IBIS), Québec City, QC, Canada; Centre de recherche sur la fonction, la structure et l'ingénierie des protéines (PROTEO),Université Laval, Québec City, QC, Canada
| | - Patrick Lagüe
- Centre de recherche sur la fonction, la structure et l'ingénierie des protéines (PROTEO),Université Laval, Québec City, QC, Canada; Département de biochimie microbiologie et bio-informatique, Institut de biologie intégrative et des systèmes (IBIS), Québec City, QC, Canada
| | - Martin Perreault
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Jean-François Thériault
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Sheng-Xiang Lin
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Donald Poirier
- Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4), Québec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
40
|
Zheng N, Liu L, Liu W, Zhang P, Huang H, Zang L, Hayashi T, Tashiro SI, Onodera S, Xia M, Ikejima T. ERβ up-regulation was involved in silibinin-induced growth inhibition of human breast cancer MCF-7 cells. Arch Biochem Biophys 2016; 591:141-9. [DOI: 10.1016/j.abb.2016.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/30/2015] [Accepted: 01/03/2016] [Indexed: 12/11/2022]
|
41
|
Zhao Z, Wang L, James T, Jung Y, Kim I, Tan R, Hoffmann FM, Xu W. Reciprocal Regulation of ERα and ERβ Stability and Activity by Diptoindonesin G. ACTA ACUST UNITED AC 2015; 22:1608-21. [PMID: 26670079 DOI: 10.1016/j.chembiol.2015.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/01/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
ERβ is regarded as a "tumor suppressor" in breast cancer due to its anti-proliferative effects. However, unlike ERα, ERβ has not been developed as a therapeutic target in breast cancer due to loss of ERβ in aggressive cancers. In a small-molecule library screen for ERβ stabilizers, we identified Diptoindonesin G (Dip G), which significantly increases ERβ protein stability while decreasing ERα protein levels. Dip G enhances the transcription and anti-proliferative activities of ERβ, while attenuating the transcription and proliferative effects of ERα. Further investigation revealed that instead of targeting ER, Dip G targets the CHIP E3 ubiquitin ligase shared by ERα and ERβ. Thus, Dip G is a dual-functional moiety that reciprocally controls ERα and ERβ protein stability and activities via an indirect mechanism. The ERβ stabilization effects of Dip G may enable the development of ERβ-targeted therapies for human breast cancers.
Collapse
Affiliation(s)
- Zibo Zhao
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Lu Wang
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Taryn James
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Youngeun Jung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406-840, Republic of Korea
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406-840, Republic of Korea
| | - Renxiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, Nanjing University, Nanjing 210093, PR China
| | - F Michael Hoffmann
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
42
|
Beekmann K, de Haan LHJ, Actis-Goretta L, Houtman R, van Bladeren PJ, Rietjens IMCM. The effect of glucuronidation on isoflavone induced estrogen receptor (ER)α and ERβ mediated coregulator interactions. J Steroid Biochem Mol Biol 2015; 154:245-53. [PMID: 26361015 DOI: 10.1016/j.jsbmb.2015.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/07/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023]
Abstract
Non-prenylated isoflavone aglycones are known to have phyto-estrogenic properties and act as agonistic ligands on ERα and ERβ due to their structural resemblance to 17β-estradiol (E2). Genistein and daidzein are the two main dietary isoflavones; upon uptake they are extensively metabolized and exist nearly exclusively as their conjugated forms in biological fluids. Little is known about the effect of conjugation on the intrinsic estrogenic activities of these isoflavones. To characterize and compare the intrinsic estrogenic activities of genistein and daidzein, and their respective 7-O-glucuronide metabolites a cell-free assay system was employed that determines the ligand-induced changes in ERα- and ERβ-ligand binding domain (LBD) interactions with 154 different binding motifs derived from 66 different nuclear receptor coregulators. The glucuronides were 8 to 4400 times less potent than their respective aglycones to modulate ERα-LBD and ERβ-LBD-coregulator interactions. Glucuronidation changed the preferential activation of genistein from ERβ-LBD to ERα-LBD and further increased the slightly preferential activation of daidzein for ERα-LBD. The tested isoflavone compounds were less potent than E2 (around 5 to 1580 times for the aglycones) but modulated the LBD-coregulator interactions in a manner similar to E2. Our results show that genistein and daidzein remain agonistic ligands of ERα-LBD and ERβ-LBD in their conjugated form with a higher relative preference for ERα-LBD than the corresponding aglycones. This shift in receptor preference is of special interest as the preferential activation of ERβ is considered one of the possible modes of action underlying the supposed beneficial instead of adverse health effects of isoflavones.
Collapse
Affiliation(s)
- Karsten Beekmann
- Division of Toxicology, Wageningen University, Postbus 8000, Bode 92, 6700EA Wageningen, the Netherlands.
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University, Postbus 8000, Bode 92, 6700EA Wageningen, the Netherlands
| | - Lucas Actis-Goretta
- Nestlé Research Center, Nestec Ltd., Vers-chez-les-Blanc, Case Postale 44, 1000 Lausanne 26, Switzerland
| | - René Houtman
- PamGene International B.V., Wolvenhoek 10, 5211HH 's-Hertogenbosch, the Netherlands
| | - Peter J van Bladeren
- Division of Toxicology, Wageningen University, Postbus 8000, Bode 92, 6700EA Wageningen, the Netherlands; Nestlé Research Center, Nestec Ltd., Vers-chez-les-Blanc, Case Postale 44, 1000 Lausanne 26, Switzerland
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Postbus 8000, Bode 92, 6700EA Wageningen, the Netherlands
| |
Collapse
|
43
|
Antiproliferative and Antiestrogenic Activities of Bonediol an Alkyl Catechol from Bonellia macrocarpa. BIOMED RESEARCH INTERNATIONAL 2015; 2015:847457. [PMID: 26557704 PMCID: PMC4628711 DOI: 10.1155/2015/847457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/24/2015] [Accepted: 09/30/2015] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to investigate antiproliferative activity of bonediol, an alkyl catechol isolated from the Mayan medicinal plant Bonellia macrocarpa. Bonediol was assessed for growth inhibition of androgen-sensitive (LNCaP), androgen-insensitive (PC-3), and metastatic androgen-insensitive (PC-3M) human prostate tumor cells; toxicity on normal cell line (HEK 293) was also evaluated. Hedgehog pathway was evaluated and competitive 3H-estradiol ligand binding assay was performed. Additionally, antioxidant activity on Nrf2-ARE pathway was evaluated. Bonediol induced a growth inhibition on prostate cancer cell lines (IC50 from 8.5 to 20.6 µM). Interestingly, bonediol binds to both estrogen receptors (ERα (2.5 µM) and ERβ (2.1 µM)) and displaces the native ligand E2 (17β-estradiol). No significant activity was found in the Hedgehog pathway. Additionally, activity of bonediol on Nrf2-ARE pathway suggested that bonediol could induce oxidative stress and activation of detoxification enzymes at 1 µM (3.8-fold). We propose that the compound bonediol may serve as a potential chemopreventive treatment with therapeutic potential against prostate cancer.
Collapse
|
44
|
Hieken TJ, Carter JM, Hawse JR, Hoskin TL, Bois M, Frost M, Hartmann LC, Radisky DC, Visscher DW, Degnim AC. ERβ expression and breast cancer risk prediction for women with atypias. Cancer Prev Res (Phila) 2015; 8:1084-92. [PMID: 26276747 DOI: 10.1158/1940-6207.capr-15-0198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/03/2015] [Indexed: 11/16/2022]
Abstract
Estrogen receptor (ER) β is highly expressed in normal breast epithelium and a putative tumor suppressor. Atypical hyperplasia substantially increases breast cancer risk, but identification of biomarkers to further improve risk stratification is needed. We evaluated ERβ expression in breast tissues from women with atypical hyperplasia and association with subsequent breast cancer risk. ERβ expression was examined by immunohistochemistry in a well-characterized 171-women cohort with atypical hyperplasia diagnosed 1967-1991. Nuclear ERβ percent and intensity was scored in the atypia and adjacent normal lobules. An ERβ sum score (percent + intensity) was calculated and grouped as low, moderate, or high. Competing risks regression was used to assess associations of ERβ expression with breast cancer risk. After 15-year median follow-up, 36 women developed breast cancer. ERβ expression was lower in atypia lobules in than normal lobules, by percent staining and intensity (both P < 0.001). Higher ERβ expression in the atypia or normal lobules, evaluated by percent staining, intensity or sum score, decreased the risk of subsequent breast cancer by 2-fold (P = 0.04) and 2.5-fold (P = 0.006). High normal lobule ERβ expression conferred the strongest protective effect in premenopausal women: the 20-year cumulative incidence of breast cancer was 0% for women younger than 45 years with high versus 31% for low-moderate ERβ expression (P = 0.0008). High ERβ expression was associated with a significantly decreased risk of breast cancer in women with atypical hyperplasia. These data suggest that ERβ may be a useful biomarker for risk stratification and a novel therapeutic target for breast cancer risk reduction.
Collapse
Affiliation(s)
- Tina J Hieken
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Tanya L Hoskin
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Melanie Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Marlene Frost
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Derek C Radisky
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
45
|
Islam MA, Bekele R, Vanden Berg JHJ, Kuswanti Y, Thapa O, Soltani S, van Leeuwen FXR, Rietjens IMCM, Murk AJ. Deconjugation of soy isoflavone glucuronides needed for estrogenic activity. Toxicol In Vitro 2015; 29:706-15. [PMID: 25661160 DOI: 10.1016/j.tiv.2015.01.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 01/17/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
Abstract
Soy isoflavones (SIF) are present in the systemic circulation as conjugated forms of which the estrogenic potency is not yet clear. The present study provides evidence that the major SIF glucuronide metabolites in blood, genistein-7-O-glucuronide (GG) and daidzein-7-O-glucuronide (DG), only become estrogenic after deconjugation. The estrogenic potencies of genistein (Ge), daidzein (Da), GG and DG were determined using stably transfected U2OS-ERα, U2OS-ERβ reporter gene cells and proliferation was tested in T47D-ERβ cells mimicking the ERα/ERβ ratio of healthy breast cells and inT47D breast cancer cells. In all assays applied, the estrogenic potency of the aglycones was significantly higher than that of their corresponding glucuronides. UPLC analysis revealed that in U2OS and T47D cells, 0.2-1.6% of the glucuronides were deconjugated to their corresponding aglycones. The resulting aglycone concentrations can account for the estrogenicity observed upon glucuronide exposure. Interestingly, under similar experimental conditions, rat breast tissue S9 fraction was about 30 times more potent in deconjugating these glucuronides than human breast tissue S9 fraction. Our study confirms that SIF glucuronides are not estrogenic as such, and that the small % of deconjugation in the cell is enough to explain the slight bioactivity observed for the SIF-glucuronides. Species differences in deconjugation capacity should be taken into account when basing risk-benefit assessment of these SIF for the human population on animal data.
Collapse
Affiliation(s)
- M A Islam
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands.
| | - R Bekele
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - J H J Vanden Berg
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - Y Kuswanti
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - O Thapa
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - S Soltani
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - F X R van Leeuwen
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - I M C M Rietjens
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| | - A J Murk
- Division of Toxicology, Wageningen University, 6703 HE Wageningen, The Netherlands
| |
Collapse
|
46
|
Hamilton N, Márquez-Garbán D, Mah V, Elshimali Y, Elashoff D, Garon E, Vadgama J, Pietras R. Estrogen Receptor-β and the Insulin-Like Growth Factor Axis as Potential Therapeutic Targets for Triple-Negative Breast Cancer. Crit Rev Oncog 2015; 20:373-90. [PMID: 27279236 PMCID: PMC5495464 DOI: 10.1615/critrevoncog.v20.i5-6.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancers (TNBCs) lack estrogen receptor-α (ERα), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) amplification and account for almost half of all breast cancer deaths. This breast cancer subtype largely affects women who are premenopausal, African-American, or have BRCA1/2 mutations. Women with TNBC are plagued with higher rates of distant metastasis that significantly diminish their overall survival and quality of life. Due to their poor response to chemotherapy, patients with TNBC would significantly benefit from development of new targeted therapeutics. Research suggests that the insulin-like growth factor (IGF) family and estrogen receptor beta-1 (ERβ1), due to their roles in metabolism and cellular regulation, might be attractive targets to pursue for TNBC management. Here, we review the current state of the science addressing the roles of ERβ1 and the IGF family in TNBC. Further, the potential benefit of metformin treatment in patients with TNBC as well as areas of therapeutic potential in the IGF-ERβ1 pathway are highlighted.
Collapse
Affiliation(s)
- Nalo Hamilton
- UCLA School of Nursing, Los Angeles, CA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | - Diana Márquez-Garbán
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Vei Mah
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Yayha Elshimali
- Department of Medicine, Division of Cancer Research and Training, Charles Drew University School of Medicine and Science, Los Angeles, CA
| | - David Elashoff
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
- Department of Medicine, Division of General Internal Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Edward Garon
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Jaydutt Vadgama
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
- Department of Medicine, Division of Cancer Research and Training, Charles Drew University School of Medicine and Science, Los Angeles, CA
| | - Richard Pietras
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
- Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
47
|
Reese JM, Suman VJ, Subramaniam M, Wu X, Negron V, Gingery A, Pitel KS, Shah SS, Cunliffe HE, McCullough AE, Pockaj BA, Couch FJ, Olson JE, Reynolds C, Lingle WL, Spelsberg TC, Goetz MP, Ingle JN, Hawse JR. ERβ1: characterization, prognosis, and evaluation of treatment strategies in ERα-positive and -negative breast cancer. BMC Cancer 2014; 14:749. [PMID: 25288324 PMCID: PMC4196114 DOI: 10.1186/1471-2407-14-749] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/25/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The role and clinical value of ERβ1 expression is controversial and recent data demonstrates that many ERβ antibodies are insensitive and/or non-specific. Therefore, we sought to comprehensively characterize ERβ1 expression across all sub-types of breast cancer using a validated antibody and determine the roles of this receptor in mediating response to multiple forms of endocrine therapy both in the presence and absence of ERα expression. METHODS Nuclear and cytoplasmic expression patterns of ERβ1 were analyzed in three patient cohorts, including a retrospective analysis of a prospective adjuvant tamoxifen study and a triple negative breast cancer cohort. To investigate the utility of therapeutically targeting ERβ1, we generated multiple ERβ1 expressing cell model systems and determined their proliferative responses following anti-estrogenic or ERβ-specific agonist exposure. RESULTS Nuclear ERβ1 was shown to be expressed across all major sub-types of breast cancer, including 25% of triple negative breast cancers and 33% of ER-positive tumors, and was associated with significantly improved outcomes in ERα-positive tamoxifen-treated patients. In agreement with these observations, ERβ1 expression sensitized ERα-positive breast cancer cells to the anti-cancer effects of selective estrogen receptor modulators (SERMs). However, in the absence of ERα expression, ERβ-specific agonists potently inhibited cell proliferation rates while anti-estrogenic therapies were ineffective. CONCLUSIONS Using a validated antibody, we have confirmed that nuclear ERβ1 expression is commonly present in breast cancer and is prognostic in tamoxifen-treated patients. Using multiple breast cancer cell lines, ERβ appears to be a novel therapeutic target. However, the efficacy of SERMs and ERβ-specific agonists differ as a function of ERα expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 16-01B Guggenheim Building, 200 First St, SW, Rochester, MN 55905, USA.
| |
Collapse
|
48
|
Abstract
Proliferative thyroid diseases are more prevalent in females than in males. Upon the onset of puberty, the incidence of thyroid cancer increases in females only and declines again after menopause. Estrogen is a potent growth factor both for benign and malignant thyroid cells that may explain the sex difference in the prevalence of thyroid nodules and thyroid cancer. It exerts its growth-promoting effect through a classical genomic and a non-genomic pathway, mediated via a membrane-bound estrogen receptor. This receptor is linked to the tyrosine kinase signaling pathways MAPK and PI3K. In papillary thyroid carcinomas, these pathways may be activated either by a chromosomal rearrangement of the tyrosine receptor kinase TRKA, by RET/PTC genes, or by a BRAF mutation and, in addition, in females they may be stimulated by high levels of estrogen. Furthermore, estrogen is involved in the regulation of angiogenesis and metastasis that are critical for the outcome of thyroid cancer. In contrast to other carcinomas, however, detailed knowledge on this regulation is still missing for thyroid cancer.
Collapse
Affiliation(s)
- Michael Derwahl
- Department of MedicineSt Hedwig Hospital and Charite, University Medicine Berlin, Grosse Hamburger Straße 5-11, 10115 Berlin, Germany
| | - Diana Nicula
- Department of MedicineSt Hedwig Hospital and Charite, University Medicine Berlin, Grosse Hamburger Straße 5-11, 10115 Berlin, Germany
| |
Collapse
|
49
|
Evers NM, van den Berg JHJ, Wang S, Melchers D, Houtman R, de Haan LHJ, Ederveen AGH, Groten JP, Rietjens IMCM. Cell proliferation and modulation of interaction of estrogen receptors with coregulators induced by ERα and ERβ agonists. J Steroid Biochem Mol Biol 2014; 143:376-85. [PMID: 24923734 DOI: 10.1016/j.jsbmb.2014.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 05/27/2014] [Accepted: 06/06/2014] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to investigate modulation of the interaction of the ERα and ERβ with coregulators in the ligand responses induced by estrogenic compounds. To this end, selective ERα and ERβ agonists were characterized for intrinsic relative potency reflected by EC50 and maximal efficacy towards ERα and ERβ mediated response in ER selective reporter gene assays, and subsequently tested for induction of cell proliferation in T47D-ERβ cells with variable ERα/ERβ ratio, and finally for ligand dependent modulation of the interaction of ERα and ERβ with coregulators using the MARCoNI assay, with 154 unique nuclear receptor coregulator peptides derived from 66 different coregulators. Results obtained reveal an important influence of the ERα/ERβ ratio and receptor selectivity of the compounds tested on induction of cell proliferation. ERα agonists activate cell proliferation whereas ERβ suppresses ERα mediated cell proliferation. The responses in the MARCoNI assay reveal that upon ERα or ERβ activation by a specific agonist, the modulation of the interaction of the ERs with coregulators is very similar indicating only a limited number of differences upon ERα or ERβ activation by a specific ligand. Differences in the modulation of the interaction of the ERs with coregulators between the different agonists were more pronounced. Based on ligand dependent differences in the modulation of the interaction of the ERs with coregulators, the MARCoNI assay was shown to be able to classify the ER agonists discriminating between different agonists for the same receptor, a characteristic not defined by the ER selective reporter gene or proliferation assays. It is concluded that the ultimate effect of the model compounds on proliferation of estrogen responsive cells depends on the intrinsic relative potency of the agonist towards ERα and ERβ and the cellular ERα/ERβ ratio whereas differences in the modulation of the interaction of the ERα and ERβ with coregulators contribute to the ligand dependent responses induced by estrogenic compounds.
Collapse
Affiliation(s)
- Nynke M Evers
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands.
| | | | - Si Wang
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| | - Diana Melchers
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - René Houtman
- PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| | - Antwan G H Ederveen
- Pharmacokinetics Pharmacodynamics & Drug Metabolism, MSD, P.O. Box 20, 5340 BH Oss, the Netherlands
| | - John P Groten
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands; PamGene International B.V., Wolvenhoek 10, 5211 HH 's Hertogenbosch, the Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, the Netherlands
| |
Collapse
|
50
|
Martinkovich S, Shah D, Planey SL, Arnott JA. Selective estrogen receptor modulators: tissue specificity and clinical utility. Clin Interv Aging 2014; 9:1437-52. [PMID: 25210448 PMCID: PMC4154886 DOI: 10.2147/cia.s66690] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Selective estrogen receptor modulators (SERMs) are a diverse group of nonsteroidal compounds that function as agonists or antagonists for estrogen receptors (ERs) in a target gene-specific and tissue-specific fashion. SERM specificity involves tissue-specific expression of ER subtypes, differential expression of co-regulatory proteins in various tissues, and varying ER conformational changes induced by ligand binding. To date, the major clinical applications of SERMs are their use in the prevention and treatment of breast cancer, the prevention of osteoporosis, and the maintenance of beneficial serum lipid profiles in postmenopausal women. However, SERMs have also been found to promote adverse effects, including thromboembolic events and, in some cases, carcinogenesis, that have proven to be obstacles in their clinical utility. In this review, we discuss the mechanisms of SERM tissue specificity and highlight the therapeutic application of well-known and emergent SERMs.
Collapse
Affiliation(s)
- Stephen Martinkovich
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| | - Darshan Shah
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| | - Sonia Lobo Planey
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| | - John A Arnott
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| |
Collapse
|