1
|
Amendoeira AF, Luz A, Valente R, Roma-Rodrigues C, Ali H, van Lier JE, Marques F, Baptista PV, Fernandes AR. Cell Uptake of Steroid-BODIPY Conjugates and Their Internalization Mechanisms: Cancer Theranostic Dyes. Int J Mol Sci 2023; 24:3600. [PMID: 36835012 PMCID: PMC9963437 DOI: 10.3390/ijms24043600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Estradiol-BODIPY linked via an 8-carbon spacer chain and 19-nortestosterone- and testosterone-BODIPY linked via an ethynyl spacer group were evaluated for cell uptake in the breast cancer cell lines MCF-7 and MDA-MB-231 and prostate cancer cell lines PC-3 and LNCaP, as well as in normal dermal fibroblasts, using fluorescence microscopy. The highest level of internalization was observed with 11β-OMe-estradiol-BODIPY 2 and 7α-Me-19-nortestosterone-BODIPY 4 towards cells expressing their specific receptors. Blocking experiments showed changes in non-specific cell uptake in the cancer and normal cells, which likely reflect differences in the lipophilicity of the conjugates. The internalization of the conjugates was shown to be an energy-dependent process that is likely mediated by clathrin- and caveolae-endocytosis. Studies using 2D co-cultures of cancer cells and normal fibroblasts showed that the conjugates are more selective towards cancer cells. Cell viability assays showed that the conjugates are non-toxic for cancer and/or normal cells. Visible light irradiation of cells incubated with estradiol-BODIPYs 1 and 2 and 7α-Me-19-nortestosterone-BODIPY 4 induced cell death, suggesting their potential for use as PDT agents.
Collapse
Affiliation(s)
- Ana F. Amendoeira
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| | - André Luz
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| | - Ruben Valente
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| | - Catarina Roma-Rodrigues
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| | - Hasrat Ali
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H5N4, Canada
| | - Johan E. van Lier
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H5N4, Canada
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela, Portugal
| | - Pedro V. Baptista
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, 2819-516 Caparica, Portugal
| |
Collapse
|
2
|
Gai L, Sun W. Recent advances in estrogen receptor-targeted probes conjugated to BODIPY dyes. Steroids 2022; 183:109031. [PMID: 35381270 DOI: 10.1016/j.steroids.2022.109031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/19/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
Estrogens, is a class of steroid hormones associated with the occurrence and development of breast cancer, that bind to estrogen receptors (ER). The development of BODIPY-based fluorescent ligands for the ER has continued to gain tremendous attention over the past 20 years. This review focuses on the synthesis methods, optical properties, and biological activity of BODIPY fluorescent probes conjugated to ER ligands. These will provide new strategy for designing fluorescent probes for targeting estrogen receptors.
Collapse
Affiliation(s)
- Linlin Gai
- Central Laboratory, Weifang People's Hospital, Weifang, Shandong 261041, PR China.
| | - Weice Sun
- Vascular Surgery, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, PR China
| |
Collapse
|
3
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
4
|
Li Z, Jiang J, Yi X, Wang G, Wang S, Sun X. miR-18b regulates the function of rabbit ovary granulosa cells. Reprod Fertil Dev 2021; 33:363-371. [PMID: 33641714 DOI: 10.1071/rd20237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/17/2021] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) have been determined to participate in the process of oestradiol production. Generally, there are two pathways by which oestradiol levels change, one being the state of cells (i.e. the status of enzymes involved in the synthesis of hormones such as oestradiol) and the other being the number of cells that secrete oestradiol. It is known that oestrogens are the main steroids produced by granulosa cells (GCs) of mature ovarian follicles. In this study we explored the function of miR-18b in rabbit GCs by overexpressing or inhibiting its activity. We found that miR-18b silencing promoted the secretion of oestradiol by significantly affecting the expression of steroidogenesis-related genes. Thus, miR-18b may act as a negative regulator of the production of enzymes related to oestradiol synthesis and affect oestradiol production. Furthermore, the effects of miR-18b on the proliferation, cell cycle and apoptosis of GCs were investigated using a cell counting kit (CCK-8) proliferation assay, detection of annexin V-fluorescein isothiocyanate apoptosis, flow cytometry and quantitative polymerase chain reaction. The results showed that miR-18b upregulated GC apoptosis (miR-18b overexpression decreases cell growth and stimulates apoptosis). These findings suggest that miR-18b and the oestrogen receptor 1 (ESR1) gene may be attractive targets to further explore the molecular regulation of GCs. The miR-18b may also explain, in part, the abnormal folliculogenesis in mammals caused by conditions such as polycystic ovary syndrome, primary ovarian insufficiency, and others.
Collapse
Affiliation(s)
- Ze Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Junyi Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xiaohua Yi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Guoyan Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Shuhui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xiuzhu Sun
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, PR China; and Corresponding author.
| |
Collapse
|
5
|
De Vitto H, Ryu J, Calderon-Aparicio A, Monts J, Dey R, Chakraborty A, Lee MH, Bode AM, Dong Z. Estrogen-related receptor alpha directly binds to p53 and cooperatively controls colon cancer growth through the regulation of mitochondrial biogenesis and function. Cancer Metab 2020; 8:28. [PMID: 33303020 PMCID: PMC7731476 DOI: 10.1186/s40170-020-00234-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 11/30/2020] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Of the genes that control mitochondrial biogenesis and function, ERRα emerges as a druggable metabolic target to be exploited for cancer therapy. Of the genes mutated in cancer, TP53 remains the most elusive to target. A clear understanding of how mitochondrial druggable targets can be accessed to exploit the underlying mechanism(s) explaining how p53-deficient tumors promote cell survival remains elusive. METHODS We performed protein-protein interaction studies to demonstrate that ERRα binds to p53. Moreover, we used gene silencing and pharmacological approaches in tandem with quantitative proteomics analysis by SWATH-MS to investigate the role of the ERRα/p53 complex in mitochondrial biogenesis and function in colon cancer. Finally, we designed in vitro and in vivo studies to investigate the possibility of targeting colon cancers that exhibit defects in p53. RESULTS Here, we are the first to identify a direct protein-protein interaction between the ligand-binding domain (LBD) of ERRα and the C-terminal domain (CTD) of p53. ERRα binds to p53 regardless of p53 mutational status. Furthermore, we show that the ERRα and p53 complex cooperatively control mitochondrial biogenesis and function. Targeting ERRα creates mitochondrial metabolic stresses, such as production of reactive oxygen species (ROS) and mitochondrial membrane permeabilization (MMP), leading to a greater cytotoxic effect that is dependent on the presence of p53. Pharmacological inhibition of ERRα impairs the growth of p53-deficient cells and of p53 mutant patient-derived colon xenografts (PDX). CONCLUSIONS Therefore, our data suggest that by using the status of the p53 protein as a selection criterion, the ERRα/p53 transcriptional axis can be exploited as a metabolic vulnerability.
Collapse
Affiliation(s)
- Humberto De Vitto
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Ali Calderon-Aparicio
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Josh Monts
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Raja Dey
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Abhijit Chakraborty
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA
| | - Mee-Hyun Lee
- Department of Pathophysiology, Zhengzhou University School of Medicine, 40 North Road, 27 District University, Zhengzhou, 450052, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, 55912, USA.
| | - Zigang Dong
- Department of Pathophysiology, Zhengzhou University School of Medicine, 40 North Road, 27 District University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
De Vitto H, Bode AM, Dong Z. The PGC-1/ERR network and its role in precision oncology. NPJ Precis Oncol 2019; 3:9. [PMID: 30911677 PMCID: PMC6428848 DOI: 10.1038/s41698-019-0081-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Transcriptional regulators include a superfamily of nuclear proteins referred to as co-activators and co-repressors, both of which are involved in controlling the functions of several nuclear receptors (NRs). The Nuclear Receptor Signaling Atlas (NURSA) has cataloged the composition of NRs, co-regulators, and ligands present in the human cell and their effort has been identified in more than 600 potential molecules. Given the importance of co-regulators in steroid, retinoid, and thyroid hormone signaling networks, hypothesizing that NRs/co-regulators are implicated in a wide range of pathologies are tempting. The co-activators known as peroxisome proliferator-activated receptor gamma co-activator 1 (PGC-1) and their key nuclear partner, the estrogen-related receptor (ERR), are emerging as pivotal transcriptional signatures that regulate an extremely broad repertoire of mitochondrial and metabolic genes, making them very attractive drug targets for cancer. Several studies have provided an increased understanding of the functional and structural biology of nuclear complexes. However, more comprehensive work is needed to create different avenues to explore the therapeutic potential of NRs/co-activators in precision oncology. Here, we discuss the emerging data associated with the structure, function, and molecular biology of the PGC-1/ERR network and address how the concepts evolving from these studies have deepened our understanding of how to develop more effective treatment strategies. We present an overview that underscores new biological insights into PGC-1/ERR to improve cancer outcomes against therapeutic resistance. Finally, we discuss the importance of exploiting new technologies such as single-particle cryo-electron microscopy (cryo-EM) to develop a high-resolution biological structure of PGC-1/ERR, focusing on novel drug discovery for precision oncology.
Collapse
Affiliation(s)
- Humberto De Vitto
- The Hormel Institute, University of Minnesota, 801 16th Avenue, Austin, NE 55912 USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue, Austin, NE 55912 USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, 801 16th Avenue, Austin, NE 55912 USA
| |
Collapse
|
7
|
He F, Wang Z, Guo G. Postnatal separation prevents the development of prenatal stress-induced anxiety in association with changes in oestrogen receptor and oxytocin immunoreactivity in female mandarin vole (Microtus mandarinus) offspring. Eur J Neurosci 2017; 47:95-108. [PMID: 29205599 DOI: 10.1111/ejn.13788] [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: 02/06/2017] [Revised: 11/15/2017] [Accepted: 11/23/2017] [Indexed: 11/27/2022]
Abstract
Oestrogen has both anxiogenic and anxiolytic effects because of variation in opposing action on alpha (ERα) and beta (ERβ) estrogen receptors in the medial preoptic area (mPOA), bed nucleus of the stria terminalis (BNST) and medial amygdala (MeA). Oxytocin (OT) reverses some of the anxiogenic effects of oestrogen in the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON). Because anxiety disorders are twice as common in women as in men, and oestrogen and OT are more important in females, we examined interactions between prenatal restraint stress (GS) and postnatal early short-term maternal separation (MS) and female mandarin vole behaviour, estrogen receptors and OT. The results show that adult female offspring from GS/noMS mothers showed increased anxiety in open-field and elevated plus-maze tests and had lower serum 17-beta-oestradiol (E2 ) levels than female offspring from GS/MS, noGS/MS and noGS/noMS mothers. GS/noMS females had more immunoreactive neurons for ERα in several brain regions and less ERβ- and OT-immunoreactive neurons in brain areas compared to GS/MS, noGS/MS and noGS/noMS offspring. Interestingly, noGS/MS and GS/MS offspring were similar to noGS/noMS offspring in that they did not develop anxiety as adults. We propose that MS alters the serum concentration of E2 and that the ERβ/ERα ratio and OT level in the brain may be responsible for the decrease in anxiety-like behaviour in adult female offspring initially exposed to anxiety-inducing conditions via an adverse foetal environment.
Collapse
Affiliation(s)
- Fengqin He
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi, 710065, China
| | - Zijian Wang
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi, 710065, China
| | - Guanlin Guo
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi, 710065, China
| |
Collapse
|
8
|
Osati S, Ali H, Guerin B, van Lier JE. Synthesis and spectral properties of estrogen- and androgen-BODIPY conjugates. Steroids 2017; 123:27-36. [PMID: 28483507 DOI: 10.1016/j.steroids.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/12/2017] [Accepted: 04/23/2017] [Indexed: 01/02/2023]
Abstract
To develop receptor based fluorescence ligands for imaging breast and prostate cancer, a series of estrogen-, testosterone- and 19-nortestosterone conjugates linked to BODIPY (4,4-difluoro-4-bora-3a,4a-diaza-s-indacene) or aza-BODIPY, were prepared. Their synthesis involves attachment of iodo derivatives of differently substituted BODIPY and aza-BODIPY analogs to the C17α-position of the steroid moieties using either the Sonogashira coupling or Click reaction. The UV-Vis absorption spectra of the conjugates range from 500 to 710nm with fluorescence emission properties ranging from 520 to 700nm, facilitating observations in living cells and tissues. Selection of the site of substitution, as well as the type of substituents on the steroidal moiety and the use of different linkers, provides a library of fluorescing conjugates to explore the effect of structural modifications on biological properties.
Collapse
Affiliation(s)
- Samira Osati
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H5N4, Canada
| | - Hasrat Ali
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H5N4, Canada
| | - Brigitte Guerin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H5N4, Canada; Centre d'Imagerie Moléculaire de I'Université de Sherbrooke (CIMUS), CR-CHUS, 3001 12(e) Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Johan E van Lier
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H5N4, Canada; Centre d'Imagerie Moléculaire de I'Université de Sherbrooke (CIMUS), CR-CHUS, 3001 12(e) Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
9
|
Jeong J, Park S, An HT, Kang M, Ko J. Small leucine zipper protein functions as a negative regulator of estrogen receptor α in breast cancer. PLoS One 2017; 12:e0180197. [PMID: 28662179 PMCID: PMC5491147 DOI: 10.1371/journal.pone.0180197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/12/2017] [Indexed: 01/15/2023] Open
Abstract
The nuclear transcription factor estrogen receptor α (ERα) plays a critical role in breast cancer progression. ERα acts as an important growth stimulatory protein in breast cancer and the expression level of ERα is tightly related to the prognosis and treatment of patients. Small leucine zipper protein (sLZIP) functions as a transcriptional cofactor by binding to various nuclear receptors, including glucocorticoid receptor, androgen receptor, and peroxisome proliferator-activated receptor γ. However, the role of sLZIP in the regulation of ERα and its involvement in breast cancer progression is unknown. We found that sLZIP binds to ERα and represses the transcriptional activity of ERα in ERα-positive breast cancer cells. sLZIP also suppressed the expression of ERα target genes. sLZIP disrupted the binding of ERα to the estrogen response element of the target gene promoter, resulting in suppression of cell proliferation. sLZIP is a novel co-repressor of ERα, and plays a negative role in ERα-mediated cell proliferation in breast cancer.
Collapse
Affiliation(s)
- Juyeon Jeong
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Sodam Park
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Hyoung-Tae An
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Minsoo Kang
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul, South Korea
- * E-mail:
| |
Collapse
|
10
|
Pavón N, Cabrera-Orefice A, Gallardo-Pérez JC, Uribe-Alvarez C, Rivero-Segura NA, Vazquez-Martínez ER, Cerbón M, Martínez-Abundis E, Torres-Narvaez JC, Martínez-Memije R, Roldán-Gómez FJ, Uribe-Carvajal S. In female rat heart mitochondria, oophorectomy results in loss of oxidative phosphorylation. J Endocrinol 2017; 232:221-235. [PMID: 27872198 DOI: 10.1530/joe-16-0161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/21/2016] [Indexed: 01/13/2023]
Abstract
Oophorectomy in adult rats affected cardiac mitochondrial function. Progression of mitochondrial alterations was assessed at one, two and three months after surgery: at one month, very slight changes were observed, which increased at two and three months. Gradual effects included decrease in the rates of oxygen consumption and in respiratory uncoupling in the presence of complex I substrates, as well as compromised Ca2+ buffering ability. Malondialdehyde concentration increased, whereas the ROS-detoxifying enzyme Mn2+ superoxide dismutase (MnSOD) and aconitase lost activity. In the mitochondrial respiratory chain, the concentration and activity of complex I and complex IV decreased. Among other mitochondrial enzymes and transporters, adenine nucleotide carrier and glutaminase decreased. 2-Oxoglutarate dehydrogenase and pyruvate dehydrogenase also decreased. Data strongly suggest that in the female rat heart, estrogen depletion leads to progressive, severe mitochondrial dysfunction.
Collapse
Affiliation(s)
- Natalia Pavón
- Departamento de FarmacologíaInstituto Nacional de Cardiología Ignacio Chávez, México, Mexico
| | - Alfredo Cabrera-Orefice
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| | | | - Cristina Uribe-Alvarez
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| | - Nadia A Rivero-Segura
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Edgar Ricardo Vazquez-Martínez
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Eduardo Martínez-Abundis
- División Académica Multidisciplinaria de ComalcalcoUniversidad Juárez Autónoma de Tabasco, México, Mexico
| | | | - Raúl Martínez-Memije
- Departamento de Instrumentación ElectromecánicaInstituto Nacional de Cardiología Ignacio Chávez, Tlalpan DF, México, Mexico
| | | | - Salvador Uribe-Carvajal
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| |
Collapse
|
11
|
Kumar R, Yadav A, Pakrasi PL. Expression of ER-α and ER-β during peri-implantation period in uterus is essential for implantation and decidualization in golden hamster. Life Sci 2016; 170:115-122. [PMID: 27939940 DOI: 10.1016/j.lfs.2016.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/23/2016] [Accepted: 12/03/2016] [Indexed: 12/19/2022]
Abstract
AIMS The role of estrogen in embryo implantation in golden hamster (Mesocricetus auratus) is still ambiguous. In order to clarify it, we investigated the spatial distribution and expression of estrogen receptors, ER-α and ER-β in the uterus of pregnant hamster during peri-implantation period and identified the effect of estrogen receptor antagonist ICI-182,780 on the embryo implantation. MAIN METHODS We performed in vivo experiments on early pregnant hamsters involving treatment with ICI-182,780, an estrogen receptor antagonist. Immunohistochemistry, western blot analysis and quantitative PCR were employed to evaluate the spatio-temporal distribution and expression of ER-α and ER-β in the uterus of normal early pregnant and treated hamsters. KEY FINDINGS Results showed that embryo implantation was completely absent in ICI-182,780 treated uterine horn while, normal implantation occurred in control and vehicle treated horns. Both the receptors were differentially expressed in the uterus of hamster from day 1 (D1) to day7 (D7). In contrast, treated horns without any implantation site showed no trace of any receptors. Protein and mRNA expression of both the receptors were high around the day of implantation while, ER-β expression was up-regulated on D7 of embryo implantation. P value˂0.05 is considered significant. SIGNIFICANCE Spatio-temporal expression of ERs in the uterus during peri-implantation period have crucial role for endometrium receptivity and implantation in hamster. Recurrent implantation failure is the devastating problem among the desirable couple and is mainly due to defect in endometrium receptivity. This study may provide a new insight to manage the problem of idiopathic infertility.
Collapse
Affiliation(s)
- Randhir Kumar
- Embryo Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| | - Akhilesh Yadav
- Indian Institute of Vegetable Research (IIVR), Varanasi, India.
| | - P L Pakrasi
- Embryo Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
12
|
Han NR, Kim HY, Yang WM, Jeong HJ, Kim HM. Glutamic acid ameliorates estrogen deficiency-induced menopausal-like symptoms in ovariectomized mice. Nutr Res 2015; 35:774-83. [PMID: 26144993 DOI: 10.1016/j.nutres.2015.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/15/2015] [Accepted: 06/09/2015] [Indexed: 12/30/2022]
Abstract
Some amino acids are considered alternative therapies for improving menopausal symptoms. Glutamic acid (GA), which is abundant in meats, fish, and protein-rich plant foods, is known to be a neurotransmitter or precursor of γ-aminobutyric acid. Although it is unclear if GA functions in menopausal symptoms, we hypothesized that GA would attenuate estrogen deficiency-induced menopausal symptoms. The objective to test our hypothesis was to examine an estrogenic effect of GA in ovariectomized (OVX) mice, estrogen receptor (ER)-positive human osteoblast-like MG-63 cells, and ER-positive human breast cancer MCF-7 cells. The results demonstrated that administration with GA to mice suppressed body weight gain and vaginal atrophy when compared with the OVX mice. A microcomputed tomographic analysis of the trabecular bone showed increases in bone mineral density, trabecular number, and connectivity density as well as a significant decrease in total porosity of the OVX mice treated with GA. In addition, GA increased serum levels of alkaline phosphatase and estrogen compared with the OVX mice. Furthermore, GA induced proliferation and increased ER-β messenger RNA (mRNA) expression, estrogen response element (ERE) activity, extracellular signal-regulated kinase phosphorylation, and alkaline phosphatase activity in MG-63 cells. In MCF-7 cells, GA also increased proliferation, Ki-67 mRNA expression, ER-β mRNA expression, and ERE activity. Estrogen response element activity increased by GA was inhibited by an estrogen antagonist. Taken together, our data demonstrated that GA has estrogenic and osteogenic activities in OVX mice, MG-63 cells, and MCF-7 cells.
Collapse
Affiliation(s)
- Na-Ra Han
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Hee-Yun Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Woong Mo Yang
- College of Korean Medicine and Institute of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Hyun-Ja Jeong
- Department of Food Technology and Inflammatory Disease Research Center, Hoseo University, Asan, Chungcheongnam-do, 336-795, Republic of Korea.
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
13
|
Han NR, Park CL, Kim NR, Kim HY, Yoou MS, Nam SY, Moon PD, Jeong HJ, Kim HM. Protective effect of porcine placenta in a menopausal ovariectomized mouse. Reproduction 2015; 150:173-81. [PMID: 26047835 DOI: 10.1530/rep-15-0157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023]
Abstract
Menopause is a significant physiological phase that occurs as women's ovaries stop producing ovum and the production of estrogen declines. Human placenta and some amino acids are known to improve menopausal symptoms. In this study, we investigated that porcine placenta extract (PPE) and arginine (Arg), a main amino acid of PPE, would have estrogenic activities in ovariectomized (OVX) mice as a menopause mouse model, human breast cancer cell line (MCF-7) cells, and human osteoblast cell line (MG-63) cells. PPE or Arg significantly inhibited the body weight and increased the vagina weight compared to the OVX mice. PPE or Arg ameliorated the vaginal atrophy in the OVX mice. The levels of 17β-estradiol and the activities of alkaline phosphatase (ALP) were significantly increased by PPE or Arg in the serum of OVX mice. Trabecular bone parameters such as bone mineral density and porosity were also improved by PPE or Arg in the OVX mice. In the MCF-7 and MG-63 cells, PPE or Arg significantly increased the cell proliferation, estrogen receptor β mRNA expression, and estrogen-response elements luciferase activity. Finally, PPE or Arg increased the activations of ALP and extracellular signal-regulated kinase 1/2 in the MG-63 cells. These results indicate that PPE or Arg would have estrogenic and osteoblastic activity. Therefore, PPE or Arg may be useful as new pharmacological tools for treating menopausal symptoms including osteoporosis. Free Korean abstract: A Korean translation of this abstract is freely available at http://www.reproduction-online.org/content/150/3/173/suppl/DC1.
Collapse
Affiliation(s)
- Na-Ra Han
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Chan-Lee Park
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Na-Rae Kim
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Hee-Yun Kim
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Myoung-Schook Yoou
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Sun-Young Nam
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Phil-Dong Moon
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Hyun-Ja Jeong
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| | - Hyung-Min Kim
- Department of PharmacologyCollege of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of KoreaDepartment of Food TechnologyInflammatory Disease Research Center, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungcheongnam-do 336-795, Republic of Korea
| |
Collapse
|
14
|
Rovani MT, Gasperin BG, Ilha GF, Ferreira R, Bohrer RC, Duggavathi R, Bordignon V, Gonçalves PBD. Expression and molecular consequences of inhibition of estrogen receptors in granulosa cells of bovine follicles. J Ovarian Res 2014; 7:96. [PMID: 25339519 PMCID: PMC4210628 DOI: 10.1186/s13048-014-0096-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/06/2014] [Indexed: 01/31/2023] Open
Abstract
Background Estradiol (E2) receptors mediate E2 effects on cell proliferation and apoptosis under normal and pathological conditions. However, the mechanisms involved in E2 signaling are not completely understood. The objectives in this study were to evaluate the expression of estrogen receptors (ESRs) during follicular selection in cattle, and the effect of intrafollicular injection of fulvestrant (an antagonist of ESRs) on follicular development and transcript abundance in granulosa cells. Methods Granulosa cells were obtained from the two largest follicles around follicular deviation, after FSH treatment and after intrafollicular injection of fulvestrant. Ovarian follicular dynamics monitored by ultrasonography and quantitative real time PCR were used to validate the in vivo model and investigate the effects of FSH supplementation or ESR blockade on mRNA expression of estradiol-related genes. Results ESR1 and ESR2 were expressed in granulosa cells of both dominant (F1) and subordinate (F2) follicles, but their transcripts levels were higher in F1 than F2 after follicular deviation. FSH treatment maintained mRNA levels of both ESR1 and ESR2 in F2 follicles at similar levels observed in F1 follicles. Intrafollicular injection of 100 μM fulvestrant inhibited follicular growth and decreased CYP19A1 mRNA levels. Transcript levels for both ESR1 and ESR2 were not affected by fulvestrant injection. Analyses of FSH-regulated genes revealed that ESRs inhibition in the dominant follicle decreased the transcript levels of the GJA1 but not those of PRKAR2B, MRO or LRP11 genes. Conclusions Our findings indicate that: both ESR1 and ESR2 are regulated during follicular deviation and dominance in cattle and in response to FSH treatment, and ESRs are required for normal gene expression and development of the dominant follicle. Furthermore, we have validated an in vivo model to study estrogen signaling during follicular development that allows paracrine signaling between different follicular cells in a physiological endocrine environment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
15
|
Thaler JD, Achari Y, Lu T, Shrive NG, Hart DA. Estrogen receptor beta and truncated variants enhance the expression of transfected MMP-1 promoter constructs in response to specific mechanical loading. Biol Sex Differ 2014; 5:14. [PMID: 25625008 PMCID: PMC4306124 DOI: 10.1186/s13293-014-0014-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/13/2014] [Indexed: 12/22/2022] Open
Abstract
Background Joint diseases such as osteoarthritis (OA) predominantly afflict post-menopausal women, suggesting a pertinent role for female hormones. Estrogen receptor beta (ER-β) has been detected in connective tissues of the knee joint suggesting that these tissues are responsive to the hormone estrogen. Matrix metalloproteinase-1 (MMP-1) activity contributes to cartilage degradation, a key factor leading to OA development in synovial joints. Two polymorphic forms of MMP-1 exist due to a deletion/insertion of the guanine residue in the promoter, and the 2G allelic variant of MMP-1 exhibits more activity than the 1G allele. Previous studies have demonstrated that the polymorphic forms of the human MMP-1 are influenced by the modulating effects of estrogen receptor isoforms. In addition to hormonal influences, physiological factors such as altered mechanical loading are also contributory features of OA. In the present study, the combined influence of biomechanical and hormonal variables on the activity of MMP-1 isoforms was evaluated. We hypothesized that the combined effects of ER-β and sheer stress will differentially activate the two allelic forms of MMP-1 in a hormone-independent manner. Methods HIG-82 synoviocytes were transiently transfected with 1G or 2G alleles (±) ER-β and subjected to either shear or equibiaxial stress. Next, 1G/2G promoter activity was measured to determine the combined influence of physiological stimuli. Truncated ER-β constructs were used to determine the importance of different domains of ER-β on 1G/2G activation. Results The 2G allele exhibited a constitutively higher activity than the 1G allele, which was further increased when the transfected cells were subject to shear stress, but not equibiaxial stress. Moreover, the combination of ER-β and shear stress further increased the activity levels of the 1G/2G allelic variants. Additionally, select AF-2 truncated ER-β variants led to increased activity levels for the 2G allele, indicating the AF-1 domain was likely involved in the response to mechanical stimulation. Conclusions These results suggest that the 1G/2G alleles of MMP-1 are influenced by specific mechanical stimuli like shear stress, as well as the ER-β receptor. These findings contribute to the potential allelic involvement in connective tissue diseases such as OA in females compared to males.
Collapse
Affiliation(s)
- John D Thaler
- McCaig Institute for Bone and Joint Health, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4 N1, AB, Canada
| | - Yamini Achari
- McCaig Institute for Bone and Joint Health, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4 N1, AB, Canada
| | - Ting Lu
- McCaig Institute for Bone and Joint Health, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4 N1, AB, Canada
| | - Nigel G Shrive
- McCaig Institute for Bone and Joint Health, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4 N1, AB, Canada ; Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary T2N 1 N4, AB, Canada
| | - David A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4 N1, AB, Canada
| |
Collapse
|
16
|
Liu HG, Li HM, Wang SY, Huang LB, Guo HJ. Gene cloning, homology comparison and analysis of the main functional structure domains of beta estrogen receptor in Jining Gray goat. Comput Biol Chem 2014; 51:42-50. [DOI: 10.1016/j.compbiolchem.2014.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 02/20/2014] [Accepted: 04/27/2014] [Indexed: 11/25/2022]
|
17
|
Verderame M, Angelini F, Limatola E. Expression of estrogen receptor alpha switches off secretory activity in the epididymal channel of the lizard Podarcis sicula. Mol Reprod Dev 2011; 79:107-17. [DOI: 10.1002/mrd.22005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 11/13/2011] [Indexed: 01/05/2023]
|
18
|
Juhasz-Böss I, Fischer C, Lattrich C, Skrzypczak M, Malik E, Ortmann O, Treeck O. Endometrial expression of estrogen receptor β and its splice variants in patients with and without endometriosis. Arch Gynecol Obstet 2010; 284:885-91. [PMID: 21110202 DOI: 10.1007/s00404-010-1768-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2010] [Accepted: 11/08/2010] [Indexed: 11/25/2022]
Abstract
BACKGROUND The role of estrogen receptor beta (ERβ) in pathogenesis of endometriosis remains to be elucidated. In this study, we have examined the expression of the four main ERβ transcript isoforms in human endometrial tissue in women with or without endometriosis. METHODS Total RNA was isolated from native endometrial tissue and transcript levels of ERα, β1, β2, β4, β5 were analyzed by means of RT-PCR. We compared the results with regard to menstrual cycle phase as well as to presence or absence of endometriosis. We prospectively harvested the endometrium of ten women without endometriosis (five for each cycle phase) and eight patients with endometriosis (five in the proliferative phase, three in the secretory phase). RESULTS ERα, β1, β2, and β5 transcripts were detected in both cycle phases. During the proliferative phase, healthy women had a significantly higher ERα/ERβ1-ratio than patients with endometriosis. Irrespective of the cycle phase, ERα-mRNA level was significantly higher than transcript levels of ERβ isoforms. CONCLUSIONS ERα, β1, β2, and β5 are expressed in human endometrium. The individual receptors differed in terms of expression strength but there was no relevant change during the cycle. The decreased ERα/ERβ1-ratio in proliferative endometrium of endometriosis patients suggest that ERβ1 might be involved in the pathogenesis of endometriosis. Further studies should be undertaken to substantiate the role of ERβ in endometrial pathology.
Collapse
Affiliation(s)
- Ingolf Juhasz-Böss
- Department of Obstetrics and Gynecology, University of Regensburg, Landshuterstr. 53, 93053 Regensburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Sanchez M, Picard N, Sauvé K, Tremblay A. Challenging estrogen receptor beta with phosphorylation. Trends Endocrinol Metab 2010; 21:104-10. [PMID: 19837602 DOI: 10.1016/j.tem.2009.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 09/23/2009] [Accepted: 09/25/2009] [Indexed: 02/07/2023]
Abstract
From classical gland-based endocrinology to nuclear hormone receptor biology, tremendous progress has been made in our understanding of hormone responses underlying cellular communication. Estrogen elicits a myriad of biological processes in reproductive and peripheral target tissues through its interaction with the estrogen receptors ERalpha and ERbeta. However, our knowledge of estrogen-dependent and independent action has mainly focused on ERalpha, leaving the role of ERbeta obscure. This review discusses our current understanding of ERbeta function and the emerging role of intracellular signals that act upon and achieve estrogen-like effects through phosphorylation of ERbeta protein. Improving our understanding of how cellular determinants impact estrogen receptor actions will likely lead to treatment strategies for related endocrine diseases affecting women's health.
Collapse
Affiliation(s)
- Mélanie Sanchez
- Department of Biochemistry, Ste-Justine Hospital Research Center, University of Montreal, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
20
|
Nakamura K, Kariyazono H. Influence of Endocrine-disrupting Chemicals on the Immune System. ACTA ACUST UNITED AC 2010. [DOI: 10.1248/jhs.56.361] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kazuo Nakamura
- Department of Biopharmaceutics, Nihon Pharmaceutical University
| | | |
Collapse
|
21
|
Mauro L, Pellegrino M, Lappano R, Vivacqua A, Giordano F, Palma MG, Andò S, Maggiolini M. E-cadherin mediates the aggregation of breast cancer cells induced by tamoxifen and epidermal growth factor. Breast Cancer Res Treat 2009; 121:79-89. [PMID: 19593637 DOI: 10.1007/s10549-009-0456-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Accepted: 06/25/2009] [Indexed: 01/03/2023]
Abstract
In the present study, we evaluated the ability of 4-hydroxytamoxifen (OHT) and epidermal growth factor (EGF) to regulate homotypic adhesion in MCF7 breast cancer cells. Our results demonstrate that OHT and EGF activate the E-cadherin promoter, increase E-cadherin mRNA and protein expression and enhance homotypic aggregation of MCF7 cells. Interestingly, an ERalpha and EGFR cross-talk is involved in the E-cadherin expression by OHT and EGF, as demonstrated by knocking down either receptor. On the basis of our findings, the well-established cross-talk between ERalpha and EGFR could be extended to the modulation of E-cadherin expression by OHT and EGF. Thus, the potential ability of tamoxifen to induce cell-cell aggregation may contribute to the biologic response of pharmacologic intervention in patients with breast cancer.
Collapse
Affiliation(s)
- Loredana Mauro
- Department Cellular Biology, University of Calabria, Cosenza, Arcavacata-Rende (CS), 87030, Italy
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
In humans, structural and functional changes attributable to aging are more visibly evident in the skin than in any other organ. Estrogens have significant effects on skin physiology and modulate epidermal keratinocytes, dermal fibroblasts and melanocytes, in addition to skin appendages including the hair follicle and the sebaceous gland. Importantly, skin aging can be significantly delayed by the administration of estrogen. This paper reviews the effects of estrogens on skin and the mechanisms by which estrogens can alleviate the changes due to aging that occur in human skin. The relevance of estrogen replacement therapy (HRT) in postmenopausal women and the potential value of selective estrogen receptor modulators (SERMs) as a therapy for diminishing skin aging are also highlighted.
Collapse
Affiliation(s)
| | - Julie Thornton
- Cutaneous Research, Medical Biosciences, School of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
23
|
Wong CKM, Lai T, Holly JMP, Wheeler MH, Stewart CEH, Farndon JR. Insulin-like growth factors (IGF) I and II utilize different calcium signaling pathways in a primary human parathyroid cell culture model. World J Surg 2006; 30:333-45. [PMID: 16485066 DOI: 10.1007/s00268-005-0339-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND In most cell types, influx of calcium (Ca2+) induces a growth or secretory response. The opposite occurs in parathyroid (PTH), cells where there is an inverse relationship between intracellular Ca2+ concentration and PTH secretion. We have examined the effects of calcium channel and metabolism modulators on insulin-like growth factors (IGFs) in a parathyroid cell culture model. METHODS Cell cultures were prepared from 9 patients undergoing operation for hyperparathyroidism. Following adhesion, the cells were transferred to serum-free medium and dosed with IGF I, II +/- ethyleneglycol-bis(beta-aminoethyl)-N,N,N',N'-tetraacetic acid (EGTA), nifedipine, nickel, 2-aminoethoxy-diphenylborate (2-APB), or dantrolene. Proliferation (96 hours) was assessed by measuring tritiated thymidine incorporation and PTH release (1 and 3 hours) assayed by IRMA. RESULTS Both IGF I and II increased DNA synthesis to 162.8% +/- 10.6% (SEM) and 131.1% +/- 7.7%, respectively (P < 0.05). EGTA at 0.2 mmol (ionized Ca2+ 0.2 mmol) did not affect the response to both IGFs. EGTA at 2 mmol (ionized Ca2+ 0 mmol) reduced the DNA synthesis of IGF I and II to 29% and 26%, respectively (P < 0.05). Nifedipine and nickel (nonspecific Ca2+ channel blocker) were equally potent in negating the mitogenic effects of both IGFs. 2-APB (IP3R blocker) reduced the basal DNA synthesis to 51.3% +/- 8.4% but had no effect on either IGF. Dantrolene (ryanodine receptor blocker) negated IGF II induced mitogenisis (74.2% +/- 6.7%) and partially inhibited IGF I mitogenesis (123% +/- 6%) (P < 0.05). The rate of PTH secretion was greater after IGF II stimulation than after IGF I stimulation. CONCLUSIONS IGFs I and II induce mitogenesis by different calcium signaling pathways. These data suggest that parathyroid cells may utilize different calcium signaling pathways to distinguish growth factors and serum calcium changes.
Collapse
Affiliation(s)
- C K M Wong
- Department of Endocrine Surgery, Frenchay Hospital, Frenchay Park, Bristol, BS16 1LE, United Kingdom.
| | | | | | | | | | | |
Collapse
|
24
|
Huang EJ, Wu CC, Lee SD, Chen JH, Liu JY, Ko JL, Lin JA, Lu MC, Chen LM, Huang CY, Kuo WW. Opposing action of estrogen receptors alpha and beta on tumor necrosis factor-alpha gene expression and caspase-8-mediated apoptotic effects in HA22T cells. Mol Cell Biochem 2006; 287:137-45. [PMID: 16633737 DOI: 10.1007/s11010-005-9092-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2005] [Accepted: 11/28/2005] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC), the major manifestation of primary liver cancer, is one of the most frequent and malignant cancers worldwide, especially in Taiwan. Estrogen receptors (ERs) have been reported to play either a proliferation- or apoptosis-enhancing role in the differentiation of cancers, including HCC. In a previous experiment, we showed that transient overexpressed estrogen receptor-alpha induced early stage HCC cell line Hep 3B cell apoptosis by increasing the hTNF-alpha gene expression in a ligand-independent manner. To further clarify if the apoptotic effect occurs in poorly differentiated HCC cell line, HA22T, and elucidate the roles of ERs and TNF-alpha, DNA fragmentation and caspase activity were measured in late stage HCC cell line, HA22T, by measuring the expression of hER-alpha and hER-beta using a Tetracycline-inducible system (Tet-on). Increased DNA fragmentation and caspase-3 activity were found in hERbeta-overexpressed HA22T cells treated with estrogen (10(-8) M) but not in hERalpha-overexpressed HA22T cells. Using RT-PCR/PCR and western blotting in HA22T cells, overexpressed hER-beta was also found to increase the expression of hTNF-alpha mRNA and induce hTNF-alpha-dependent luciferase activity in a ligand-dependent manner. Additionally, LPS treatment and hER-beta overexpression both enhance caspase-8 activities, whereas neither hER-beta nor E2 treatment affected caspase-9 activities. In addition, the overexpressed hER-beta plus E2 enhanced DNA fragmentation and caspase-8 activities were only partially reduced by anti-hTNF-alpha (0.1 ng/ml), which was possibly due to the involvement of P53 and TGF-beta. Taken together, our data indicates that overexpressed hER-beta but not hER-alpha may induce caspase-8-mediated apoptosis by increasing the hTNF-alpha gene expression in a ligand-dependent manner in poorly differentiated HA22T cells.
Collapse
Affiliation(s)
- Erh-Jung Huang
- Center of General Education, Central Taiwan University of Science & Technology, Taichung, 406, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hsu HH, Cheng SF, Chen LM, Liu JY, Chu CH, Weng YJ, Li ZY, Lin CS, Lee SD, Kuo WW, Huang CY. Over-expressed estrogen receptor-alpha up-regulates hTNF-alpha gene expression and down-regulates beta-catenin signaling activity to induce the apoptosis and inhibit proliferation of LoVo colon cancer cells. Mol Cell Biochem 2006; 289:101-9. [PMID: 16628468 DOI: 10.1007/s11010-006-9153-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Accepted: 02/10/2006] [Indexed: 12/11/2022]
Abstract
Epidemiologic studies reported that the prevalence of hereditary non-polyposis colon cancer (HNPCC) in male is about 1.5-fold higher than that in female. Decreases in circulatory estrogen (E(2)) have been reported to downregulate the expression of E(2) receptor (ER) and significantly increase the risk of colorectal cancer. Patients that received E(2) replacement therapy were found to have a reduction in the incidence of colon adenoma and carcinoma. Furthermore, significant decreases in the expression of ER have been found in colorectal cancer specimens. Evidences strongly suggest the protective roles of E(2) and ER against colorectal cancer. However, the mechanisms of ERalpha effects on colorectal cancer cells remained un-clear. LoVo cells were transient transfected to overexpress ERalpha, DNA fragmentation and the activated caspases measurements were performed to evaluate apoptotic effects. Western blotting was used to evaluate protein levels, and luciferase activity assay to measure the Htnf-a promoter activity. The results clearly demonstrated that overexpressed ERalpha with or without E(2) (10(-8) M) treatment could activate caspase -8, -9, and 3 and induce DNA fragmentation in LoVo cell. At the same time, overexpressed ERalpha plus E(2) significantly increases the expression and promoter activity of hTNF-alpha, and the DNA fragmentation effect induced by E(2) plus ERalpha were reduced by the addition of hTNF antibody (0.1 ng(ml). In addition, E(2) plus ERalpha significantly upregulated p21 and p27 levels and downregulated the beta-catenin and its target genes, cyclin D1 and Rb, which regulate the cell cycle and cell proliferation. The results indicate that E(2) plus overexpressed ERalpha induce LoVo cell apoptosis might mediate through the increase of hTNF-alpha gene expression, which in turn activate caspase-8, -9 and caspase-3 and lead to the DNA fragmentation and apoptosis. E(2) plus ERalpha also showed the downregulation of beta-catenin signalings implicating the suppression of proliferation and metastasis of colorectal cells. Efforts aiming at enhancing ERalpha expression and(or activity may be proved to be an alternative therapy against colorectal cancer.
Collapse
Affiliation(s)
- Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shi SF, Yu CQ. [Progress in research on phytoestrogens and their effect targets]. ACTA ACUST UNITED AC 2006; 3:408-10. [PMID: 16159582 DOI: 10.3736/jcim20050521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shu-Fang Shi
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | | |
Collapse
|
27
|
St-Laurent V, Sanchez M, Charbonneau C, Tremblay A. Selective hormone-dependent repression of estrogen receptor beta by a p38-activated ErbB2/ErbB3 pathway. J Steroid Biochem Mol Biol 2005; 94:23-37. [PMID: 15862947 DOI: 10.1016/j.jsbmb.2005.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Deregulated signaling of ErbB2 receptor tyrosine kinase is often associated with hormone resistance in estrogen receptor alpha (ERalpha)-positive breast cancers, establishing a relationship between ErbB2 and ERalpha pathways. Although ERalpha and ERbeta are expressed in many breast cancer cells, the response of ERbeta to ErbB2 signaling is less well defined. In the present study, we demonstrate that ERbeta activity can be modulated by ErbB2 signaling in ER-expressing breast cancer cells. The estrogen-dependent transcriptional activity of ERbeta was altered in a manner similar to ERalpha by either activation of ErbB2/ErbB3 signaling by growth factor heregulin beta or expression of a constitutively active mutant of ErbB2. However, as opposed to ERalpha, the p38 MAPK pathway was found to be involved in liganded ERbeta repression activity by ErbB2 signaling and in regulating estrogen-responsive promoter occupancy by ERbeta. The repression in ERbeta response to hormone was dependent upon its AF-1 domain which includes serines 106 and 124, two phosphorylation target sites for Erk that we previously showed to be involved in SRC-1 recruitment to ERbeta. Substitution of these two serines by aspartic acid residues abolished the repression of ERbeta by activated ErbB2/ErbB3. Moreover, expression of SRC-1 also relieved the inhibition of ERbeta in heregulin-treated cells. Our study demonstrates a functional coupling between ERbeta and ErbB receptors and outlines the differential role of the AF-1 region in the regulation of the estrogen-dependent cell growth and activity of both estrogen receptors in response to growth factor signaling.
Collapse
Affiliation(s)
- Véronique St-Laurent
- Ste-Justine Hospital Research Center, 3175 Cote Ste-Catherine, Montreal, Que., Canada H3T 1C5
| | | | | | | |
Collapse
|
28
|
Couse JF, Korach KS. Estrogen receptor-α mediates the detrimental effects of neonatal diethylstilbestrol (DES) exposure in the murine reproductive tract. Toxicology 2004; 205:55-63. [PMID: 15458790 DOI: 10.1016/j.tox.2004.06.046] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
It is generally believed that estrogen receptor-dependent and -independent pathways are involved in mediating the developmental effects of the synthetic estrogen, diethylstilbestrol (DES). However, the precise role and extent to which each pathway contributes to the resulting pathologies remains unknown. We have employed the estrogen receptor knockout (ERKO) mice, which lack either estrogen receptor-alpha (alphaERKO or estrogen receptor-beta (betaERKO), to gain insight into the contribution of each ER-dependent pathway in mediating the effects of neonatal DES exposure in the female and male reproductive tract tissues of the mouse. Estrogen receptor-alpha female mice exhibited complete resistance to the chronic effects of neonatal DES exposure that were obvious in exposed wild-type animals, including atrophy and epithelial squamous metaplasia in the uterus; proliferative lesions of the oviduct; and persistent cornification of the vaginal epithelium. DES-mediated reduction in uterine Hoxa10, Hoxa11 and Wnt7a expression that occurs wild-type females during the time of exposure was also absent in alphaERKO females. In the male, alphaERKO mice exhibited complete resistance to the chronic effects of neonatal DES exposure on the prostate, including decreased androgen receptor levels, epithelial hyperplasia, and increased basal cell proliferation. Although ERbeta is highly expressed in the prostate epithelium, DES-exposed betaERKO males exhibited all of the effects of neonatal DES exposure that were observed in similarly exposed wild-type males. Therefore, the lack of DES-effects on gene expression and tissue differentiation in the alphaERKO uterus and prostate provides unequivocal evidence of an obligatory role for ERalpha in mediating the detrimental actions of neonatal DES exposure in the murine reproductive tract.
Collapse
Affiliation(s)
- John F Couse
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, PO Box 12233, MD B3-02, NC 27709, USA
| | | |
Collapse
|
29
|
Gozgit JM, Nestor KM, Fasco MJ, Pentecost BT, Arcaro KF. Differential action of polycyclic aromatic hydrocarbons on endogenous estrogen-responsive genes and on a transfected estrogen-responsive reporter in MCF-7 cells. Toxicol Appl Pharmacol 2004; 196:58-67. [PMID: 15050408 DOI: 10.1016/j.taap.2003.12.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Accepted: 12/15/2003] [Indexed: 11/23/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are common environmental pollutants that have been extensively studied for multiple toxicological endpoints in both laboratory animals and humans. The purpose of this study was to investigate the estrogenicity of PAHs in the human breast cancer cell line MCF-7. We investigated 14 PAHs for their ability to bind either the estrogen receptor (ER) or the aryl hydrocarbon receptor (AhR) and to activate target gene expression. PAHs were tested in a human recombinant estrogen receptor (hrER) competitive binding assay, and in both an estrogen response element (ERE)- and xenobiotic response element (XRE)-mediated reporter gene assay. We used quantitative RT-PCR to examine selected PAHs that showed activity in the ERE reporter gene assay for their ability to upregulate estrogen-responsive genes HEM45, progesterone receptor, and pS2, and the aryl hydrocarbon-responsive CYP1A1 gene. None of the 14 PAHs bound the hrER, but five of the PAHs (anthracene, B[a]A, chrysene, B[b]F, and B[a]P) induced ER-reporter activity. This activity was dependent on the metabolism of PAHs in MCF-7 cells via the AhR pathway, which resulted in the formation of metabolites that bound the ER. None of the five PAHs that induced the ER-reporter were found to upregulate estrogen-responsive genes, yet four of the five PAHs induced AhR-dependent CYP1A1 gene expression. In contrast, a metabolite of B[a]P, 3'OH-B[a]P, and a PCB metabolite, 4'OH-2,4,6-BP, did weakly upregulate all three estrogen-responsive genes. Data from these studies indicate that induction of ER-reporter activity alone does not necessarily parallel endogenous gene transcription, and that the reporter gene assay may detect interactions that are not functional in vivo.
Collapse
Affiliation(s)
- Joseph M Gozgit
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003-9298, USA
| | | | | | | | | |
Collapse
|
30
|
Zhang JQ, Su BY, Cai WQ. Immunolocalization of estrogen receptor beta in the hypothalamic paraventricular nucleus of female mice during pregnancy, lactation and postnatal development. Brain Res 2004; 997:89-96. [PMID: 14715153 DOI: 10.1016/j.brainres.2003.10.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Previous studies have shown that estrogen receptor beta (ERbeta) is the predominant estrogen receptor in the hypothalamic paraventricular nucleus (PVN) of mouse, mediating estrogen regulation of the neuroendocrine activities of the PVN, but the exact roles that ERbeta plays in the PVN remain unclear. In this study, we used immunocytochemistry to investigate the expression of ERbeta in the maternal PVN of mice during pregnancy (pregnant days 8, 10, 12, 15 and 18), lactation (postpartum days 1, 4 and 8) as well as in the PVN of the females from postnatal days 1, 3, 5, 7, 9, 15, 30 and 70. We found out that ERbeta was predominantly localized in the magnocellular divisions of PVN. In the pregnant female brain, generally, the ERbeta was lower than that of the postnatal development, the lowest level was found at gestational days 10-12; then from gestational day 18 to postpartum day 1, it increased to higher levels, followed by a decrease from postpartum day 4. During the postnatal development, the highest level of ERbeta was found at early postnatal days (before postnatal day 15), thereafter, it decreased to a lower level. The above results indicate that circulating sex steroids may differentially regulate the expression of ERbeta in the PVN of mice. It also suggests that this receptor may play important roles in the regulation of parturition and in the development, food intake and body weight increases of the newborns by acting on the neuropeptides, which were also detected in the PVN.
Collapse
Affiliation(s)
- Ji-Qiang Zhang
- Department of Histology and Embryology, College of Medicine, Third Military Medical University, #30, Gao Tan Yan Street, Chongqing 400038, China.
| | | | | |
Collapse
|
31
|
Deblois G, Giguère V. Ligand-independent coactivation of ERalpha AF-1 by steroid receptor RNA activator (SRA) via MAPK activation. J Steroid Biochem Mol Biol 2003; 85:123-31. [PMID: 12943696 DOI: 10.1016/s0960-0760(03)00225-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Nuclear receptor coactivators are factors that enhance the transcriptional activity of the receptor. Coactivators usually work in ligand-independent and/or dependent manners by interacting with activation function-1 (AF-1) and AF-2 of the receptor, respectively. The recently characterized steroid receptor RNA activator (SRA) was cloned as an AF-1-dependent coactivator and shown to enhance the transcriptional activity of selected steroid receptors. In this work, we describe the effect of SRA on the activity of the two estrogen receptor (ER) isoforms, ERalpha and ERbeta. We show that SRA potentiates the estrogen-induced transcriptional activity of both ERalpha and ERbeta. We demonstrate that the transcriptional activity of ERalpha can be enhanced by SRA in a ligand-independent manner through the AF-1 domain. However, this AF-1-dependent effect of SRA is not observed on ERbeta, denoting the ability of SRA to mediate differential activation of ERalpha and ERbeta. The presence of an intact serine residue at position 118 (S(118)) in ERalpha AF-1 is required for coactivation of ERalpha by SRA. We also show that activation of the mitogen activated protein kinase (MAPK) induces ligand-independent coactivation of ERalpha by SRA, a mechanism that is independent of the AF-2. Finally, SRA is unable to rescue the loss of activity of the S(118) ERalpha mutant in response to H-Ras(V12), suggesting that phosphorylation of S(118) by MAPK participates in the ligand-independent effect of SRA on ERalpha.
Collapse
Affiliation(s)
- Geneviève Deblois
- Molecular Oncology Group, McGill University Health Center, Room H5-21, 687 Pine Avenue West, Montréal, Quebec, Canada H3A 1A1
| | | |
Collapse
|
32
|
Abstract
Because of recent concerns about the long-term risks of estrogen replacement therapy in postmenopausal women, there is growing interest in a group of compounds known as selective estrogen receptor modulators (SERMs). The SERMs bind to estrogen receptors and have tissue-specific effects that allow them to function as estrogen agonists in some tissues and estrogen antagonists in other tissues. There are four SERMs currently marketed in the United States. These include the triphenylethylenes--clomiphene citrate (Clomid), tamoxifen, and toremifene--and the benzothiophene, raloxifene. Clomid is used primarily in the treatment of infertility. Tamoxifen is indicated for the treatment and prevention of breast cancer. It has an estrogen antagonist effect on breast tissue, but an estrogen-like effect on lipids, bone, and the endometrium. Toremifene has an antagonist/agonist profile similar to that of tamoxifen. Raloxifene is approved for the prevention of osteoporosis in postmenopausal women. It is thought to be an estrogen antagonist on the uterus and breast tissues and an estrogen agonist with respect to bone and serum lipids.
Collapse
Affiliation(s)
- Sally G Haskell
- Section of General Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
33
|
Abstract
There is still extensive disparity in our understanding of how estrogens exert their actions, particularly in non-reproductive tissues such as the skin. Although it has been recognized for some time that estrogens have significant effects on many aspects of skin physiology and pathophysiology, studies on estrogen action in skin have been limited. However, estrogens clearly have an important function in many components of human skin including the epidermis, dermis, vasculature, hair follicle and the sebaceous, eccrine and apocrine glands, having significant roles in skin aging, pigmentation, hair growth, sebum production and skin cancer. The recent discovery of a second intracellular estrogen receptor (ERbeta) with different cell-specific roles to the classic estrogen receptor (ERalpha), and the identification of cell surface estrogen receptors, has provided further challenges to understanding the mechanism of estrogen action. It is now time to readdress many of the outstanding questions regarding the role of estrogens in skin and improve our understanding of the physiology and interaction of steroid hormones and their receptors in human skin. Not only will this lead to a better understanding of estrogen action, but may also provide a basis for further interventions in pathological processes that involve dysregulation of estrogen action.
Collapse
Affiliation(s)
- M J Thornton
- Department of Biomedical Sciences, University of Bradford, Bradford, West Yorkshire, UK.
| |
Collapse
|
34
|
Hall JM, Korach KS. Analysis of the molecular mechanisms of human estrogen receptors alpha and beta reveals differential specificity in target promoter regulation by xenoestrogens. J Biol Chem 2002; 277:44455-61. [PMID: 12200415 DOI: 10.1074/jbc.m200849200] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Most of the currently available information on the transcriptional activities of endocrine-disrupting chemicals (xenoestrogens) through estrogen receptors alpha (ERalpha) and beta (ERbeta) has been derived from transactivation studies on synthetic estrogen-responsive reporters. Thus, the ability of the xenoestrogen-liganded ERs to regulate endogenous estrogen-responsive gene expression has not been well characterized. Here, we have evaluated the activities of xenoestrogens through ERalpha and ERbeta on the vitellogenin A2 estrogen-response element (ERE) and the human pS2, lactoferrin, and complement 3 physiological target gene promoters. Using mammalian cell transient transfection assays, we found that the activities of xenoestrogens were mediated in a promoter-specific manner. For example, when bound to all ligands examined, ERalpha displayed high levels of transcription on the vitellogenin ERE and the lactoferrin promoter, but substantially lower activity on the complement 3 and pS2 promoters. However, one of the most important observations was that there were significant differences in the relative transcriptional activities of xenoestrogen-bound ERalpha and ERbeta on different promoters, suggesting that ERalpha and ERbeta make unique contributions to xenoestrogen action in target cells. When probing the molecular mechanism of the promoter-specific activities observed, we found that the transcriptional activity of the ERs correlated with the ability of each receptor to assume an active conformation on specific promoters. Taken together, the results indicate that the transcriptional activities of xenoestrogens are mediated in a promoter-specific manner and that estrogen-responsive promoters communicate differently with ERalpha and ERbeta by influencing their structures in a distinct manner that leads to diversity in their transcriptional responses.
Collapse
Affiliation(s)
- Julie M Hall
- Receptor Biology Section, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
35
|
Lewandowski S, Kalita K, Kaczmarek L. Estrogen receptor beta. Potential functional significance of a variety of mRNA isoforms. FEBS Lett 2002; 524:1-5. [PMID: 12135731 DOI: 10.1016/s0014-5793(02)03015-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recent cloning of estrogen receptor beta (ERbeta) was followed by the discovery of a variety of its isoforms. This review describes the complexity of ERbeta mRNAs in various species for which most data have been gathered so far. The most surprising finding is the great variation in isoform structure among various mammalian species. This may reflect either the fact that only a very limited number of isoforms have been described so far or between-species specificity, especially as common elements in closely related species could still be noted. Isoform variations, as detected mainly at the mRNA sequence level, should result in profound functional differences at the level of proteins as already shown in selected cases. Thus, it is proposed that the diversity of ERbeta isoforms implies a functional role of this phenomenon in cellular physiology and pathology of estrogen response.
Collapse
Affiliation(s)
- Sebastian Lewandowski
- Nencki Institute, Department of Molecular and Cellular Neurobiology, Pasteura 3, 02-093 Warsaw, Poland
| | | | | |
Collapse
|
36
|
|
37
|
Evans MJ, Lai K, Shaw LJ, Harnish DC, Chadwick CC. Estrogen receptor alpha inhibits IL-1beta induction of gene expression in the mouse liver. Endocrinology 2002; 143:2559-70. [PMID: 12072388 DOI: 10.1210/endo.143.7.8919] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogens have been suggested to modulate several inflammatory processes. Here, we show that IL-1beta treatment induced the expression of approximately 75 genes in the liver of ovariectomized mice. 17alpha-Ethinyl estradiol (EE) pretreatment reduced the IL-1beta induction of approximately one third of these genes. Estrogen receptor alpha (ERalpha) was required for this inhibitory activity, because EE inhibition of IL-1beta-stimulated gene expression occurred in ERbeta knockout mice, but not in ERalpha knockout mice. EE treatment induced expression of 40 genes, including the transcriptional repressor short heterodimer partner and prostaglandin D synthase, known modulators of nuclear factor-kappaB signaling. However, the ER agonists genistein and raloxifene both inhibited IL-1beta gene induction without stimulating the expression of prostaglandin D synthase, short heterodimer partner, or other ER-inducible genes, indicating that induction of gene expression was not required for ER inhibition of IL-1beta signaling. Finally, the ability of EE to repress IL-1beta gene induction varied among tissues. For example, EE inhibited IL-1beta induction of lipopolysaccharide-induced c-x-c chemokine (LIX) in the liver, but not in the spleen or lung. The degree of EE repression did not correlate with ER expression. cAMP response element binding protein-binding protein (CBP)/p300 levels also varied between tissues. Together, these results are consistent with a model of in vivo ER interference with IL-1beta signaling through a coactivator-based mechanism.
Collapse
Affiliation(s)
- Mark J Evans
- Wyeth Research, Collegeville, Pennsylvania 19426, USA.
| | | | | | | | | |
Collapse
|
38
|
Zhang JQ, Cai WQ, Zhou DS, Su BY. Distribution and differences of estrogen receptor beta immunoreactivity in the brain of adult male and female rats. Brain Res 2002; 935:73-80. [PMID: 12062475 DOI: 10.1016/s0006-8993(02)02460-5] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Studies have shown that estrogen plays important roles in regulating neural structure and function in the brain, but the mechanism remains unclear. The actions of estrogen were thought to be mediated by a single estrogen receptor until the identification of another estrogen receptor, namely estrogen receptor-beta (ER-beta). Here we report a comprehensive study of the localization of ER-beta immunoreactivity and differences in the brains of adult male and female rats on the basis of a nickel ammonium sulfate-enhanced immunocytochemical method using a polyclonal antiserum sc-8974. The results of these studies revealed: (1) ER-beta immunoactive material was mainly localized in the neuronal nucleus, but it was also detectable in the cytoplasm and neuronal processes; (2) in both male and female rats, high levels of ER-beta immunopositive signals were detected in the anterior olfactory nucleus, cerebral cortex, Purkinje cells, vertical limb of the diagonal band, red nucleus, locus ceruleus, and motor trigeminal nucleus. Moderate levels were found in the medial septum, lateral amygdaloid nucleus, substantia nigra, and central gray. Weak signals were localized in other subregions of the hypothalamus and amygdaloid complex; (3) there was an obvious difference of ER-beta immunoreactivity between male and female rats, and its intracellular distribution also showed a sex difference. The above results provide the first detailed evidence that ER-beta protein is widely distributed in both male and female rat brains, but that distinctive sex differences also exist. Estrogen may exert its function in different brain regions in a gender-specific manner.
Collapse
Affiliation(s)
- Ji Qiang Zhang
- Department of Histology and Embryology, College of Medicine, Third Military Medical University, Chongqing 400038, China.
| | | | | | | |
Collapse
|
39
|
Yi P, Driscoll MD, Huang J, Bhagat S, Hilf R, Bambara RA, Muyan M. The effects of estrogen-responsive element- and ligand-induced structural changes on the recruitment of cofactors and transcriptional responses by ER alpha and ER beta. Mol Endocrinol 2002; 16:674-93. [PMID: 11923465 DOI: 10.1210/mend.16.4.0810] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Estrogen signaling is mediated by ER alpha and -beta. ERs are converted from an inactive form to a transcriptionally active state through conformational changes induced by ligand and estrogen-responsive element (ERE) sequences. We show here that ER alpha and ER beta bind to an ERE independently from ER ligands. We found that although the binding affinity of ER beta for an ERE is 2-fold lower than that of ER alpha, both ERs use the same nucleotides for DNA contacts. We show that both EREs and ligands are independent modulators of ER conformation. Specifically, the ERE primarily determines the receptor-DNA affinity, whereas the structure of the ER ligand dictates the affinity of ER for particular cofactors. We found that the ligand-dependent cofactor transcriptional intermediary factor-2, through a distinct surface, also interacts with ER alpha preferentially and independently of ligand. The extent of interaction, however, is dependent upon the ER-ERE affinity. In transfected cells, ER alpha is more transcriptionally active than ER beta. The ERE sequence, however, determines the potency of gene induction when either ER subtype binds to an agonist. Antagonists prevent ERs from inducing transcription independently from ERE sequences. Thus, ERE- and ligand-induced structural changes are independent determinants for the recruitment of cofactors and transcriptional responses. The ability of ER alpha to differentially recruit a cofactor could contribute to ER subtype-specific gene responses.
Collapse
Affiliation(s)
- Ping Yi
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Estrogens have been shown to exert significant benefits on the cardiovascular system both in animals and in postmenopausal women. However, the exact mechanism of these effects are, for the most part, still unknown. The goal of this paper is to evaluate the role of estrogen receptors (ER) in mediating some of the cardiovascular beneficial actions of 17 beta-estradiol (E2). This analysis was possible because of the availability of ER alpha (ER alpha KO) and ER beta-deficient (ER beta KO) mice, and access to a patient with ER alpha-deficiency. Experimental results obtained in our laboratory demonstrated that the ER alpha subtype mediates E2-induced increase in endothelial nitric oxide production and facilitation of fibroblast growth factor-elicited angiogenesis in vivo. Others have confirmed these findings. Experiments using a novel ER-antagonist and ApoExER alpha double-knockout mice proved that ER alpha mediates some of the antiatherosclerotic effects of E2 as well. In contrast, both the ER alpha and ER beta subtypes appear to mediate the beneficial effects of E2 on vascular smooth muscle proliferation after vessel injury. The young male patient with ER alpha-deficiency exhibited reduced endothelial nitric oxide production and premature coronary arteriosclerosis. These studies in mice and a male human subject suggest that absence of functional ER may represent a novel risk factor for cardiovascular diseases.
Collapse
Affiliation(s)
- Gabor M Rubanyi
- Department of Gene Therapy, Berlex Biosciences, Richmond, CA, USA.
| | | | | |
Collapse
|
41
|
Monje P, Boland R. Subcellular distribution of native estrogen receptor alpha and beta isoforms in rabbit uterus and ovary. J Cell Biochem 2001; 82:467-79. [PMID: 11500923 DOI: 10.1002/jcb.1182] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The association of estrogen receptors with non-nuclear/cytoplasmic compartments in target tissues has been documented. However, limited information is available on the distribution of estrogen receptor isoforms, specially with regard to the newly described beta isotype. The subcellular localization of estrogen receptor alpha and beta isoforms was investigated in rabbit uterus and ovary. Native alpha and beta subtypes were immunodetected using specific antibodies after subjecting the tissue to fractionation by differential centrifugation. The ovary expressed alpha and beta estrogen receptors in predominant association to cytosolic components. However, in the uterus, a substantial proportion of the total estrogen binding capacity and coexpression of the two isoforms was detected in mitochondria and microsomes. The mitochondrial-enriched subfraction represented an important source of 17beta-estradiol binding, where the steroid was recognized in a stereospecific and high affinity manner. The existence of mitochondrial and membrane estrogen binding sites correlated with the presence of estrogen receptor alpha but mainly with estrogen receptor beta proteins. Using macromolecular 17beta-estradiol derivatives in Ligand Blot studies, we could confirm that both alpha and beta isoforms were expressed as the major estrogen binding proteins in the uterus, while estrogen receptor alpha was clearly the dominant isoform in the ovary. Other low molecular weight estrogen receptor alpha-like proteins were found to represent an independent subpopulation of uterine binding sites, expressed to a lesser extent. This differential cellular partitioning of estrogen receptor alpha and beta forms may contribute to the known diversity of 17beta-estradiol effects in target organs. Both estrogen receptor alpha and beta expression levels and cellular localization patterns among tissues, add complexity to the whole estrogen signaling system, in which membrane and mitochondrial events could also be implicated.
Collapse
Affiliation(s)
- P Monje
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | | |
Collapse
|
42
|
Couse JF, Dixon D, Yates M, Moore AB, Ma L, Maas R, Korach KS. Estrogen receptor-alpha knockout mice exhibit resistance to the developmental effects of neonatal diethylstilbestrol exposure on the female reproductive tract. Dev Biol 2001; 238:224-38. [PMID: 11784006 DOI: 10.1006/dbio.2001.0413] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Data indicate that estrogen-dependent and -independent pathways are involved in the teratogenic/carcinogenic syndrome that follows developmental exposure to 17beta-estradiol or diethylstilbestrol (DES), a synthetic estrogen. However, the exact role and extent to which each pathway contributes to the resulting pathology remain unknown. We employed the alphaERKO mouse, which lacks estrogen receptor-alpha (ERalpha), to discern the role of ERalpha and estrogen signaling in mediating the effects of neonatal DES exposure. The alphaERKO provides the potential to expose DES actions mediated by the second known ER, ERbeta, and those that are ER-independent. Wild-type and alphaERKO females were treated with vehicle or DES (2 microg/pup/day for Days 1-5) and terminated after 5 days and 2, 4, 8, 12, and 20 months for biochemical and histomorphological analyses. Assays for uterine expression of the genes Hoxa10, Hoxa11, and Wnt7a shortly after treatment indicated significant decreases in DES-treated wild-type but no effect in the alphaERKO. In contrast, the DES effect on uterine expression of Wnt4 and Wnt5a was preserved in both genotypes, suggesting a developmental role for ERbeta. Adult alphaERKO mice exhibited complete resistance to the chronic effects of neonatal DES exposure exhibited in treated wild-type animals, including atrophy, decreased weight, smooth muscle disorganization, and epithelial squamous metaplasia in the uterus; proliferative lesions of the oviduct; and persistent vaginal cornification. Therefore, the lack of DES effects on gene expression and tissue differentiation in the alphaERKO provides unequivocal evidence of an obligatory role for ERalpha in mediating the detrimental actions of neonatal DES exposure in the murine reproductive tract.
Collapse
Affiliation(s)
- J F Couse
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Hall JM, Couse JF, Korach KS. The multifaceted mechanisms of estradiol and estrogen receptor signaling. J Biol Chem 2001; 276:36869-72. [PMID: 11459850 DOI: 10.1074/jbc.r100029200] [Citation(s) in RCA: 825] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- J M Hall
- Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA.
| | | | | |
Collapse
|
44
|
Karas RH, Schulten H, Pare G, Aronovitz MJ, Ohlsson C, Gustafsson JA, Mendelsohn ME. Effects of estrogen on the vascular injury response in estrogen receptor alpha, beta (double) knockout mice. Circ Res 2001; 89:534-9. [PMID: 11557741 DOI: 10.1161/hh1801.097239] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The two known estrogen receptors, ERalpha and ERbeta, mediate the effects of estrogen in all target tissues, including blood vessels. We have shown previously that estrogen inhibits vascular injury response to the same extent in female wild-type (WT), ERalpha knockout (ERalphaKO(CH)), and ERbeta knockout (ERbetaKO(CH)) mice. We generated mice harboring disruptions of both ERalpha and ERbeta genes (ERalpha,betaKO(CH)) by breeding and studied the effect of 17beta-estradiol (E2) on vascular injury responses in ovariectomized female ERalpha,betaKO(CH) mice and WT littermates. E2 inhibited increases in vascular medial area following injury in the WT mice but not in the ERalpha,betaKO(CH) mice, demonstrating for the first time that the two known estrogen receptors are necessary and sufficient to mediate estrogen inhibition of a component of the vascular injury response. Surprisingly, as in WT littermates, E2 still significantly increased uterine weight and inhibited vascular smooth muscle cell (VSMC) proliferation following injury in the ERalpha,betaKO(CH) mice. These data support that the role of estrogen receptors differs for specific components of the vascular injury response in the ERalpha,betaKO(CH) mice. The results leave unresolved whether E2 inhibition of VSMC proliferation in ERalpha,betaKO(CH) mice is caused by a receptor-independent mechanism, an unidentified receptor responsive to estrogen, or residual activity of the ERalpha splice variant reported previously in the parental ERalphaKO(CH) mice. These possibilities may be resolved by studies of mice in which ERalpha has been fully disrupted (ERalphaKO(St)), which are in progress.
Collapse
Affiliation(s)
- R H Karas
- Molecular Cardiology Research Institute, Department of Medicine, Division of Cardiology, New England Medical Center Hospitals and Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Weatherman RV, Clegg NJ, Scanlan TS. Differential SERM activation of the estrogen receptors (ERalpha and ERbeta) at AP-1 sites. ACTA ACUST UNITED AC 2001; 8:427-36. [PMID: 11358690 DOI: 10.1016/s1074-5521(01)00025-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The selective estrogen receptor modulators (SERMs) raloxifene and tamoxifen are triphenylethylene derivatives that affect transcriptional regulation by the estrogen receptors (ERalpha and ERbeta) but show different effects in different tissues. A third triphenylethylene derivative, GW-5638, displays tissue selectivity in rats identical to that of raloxifene, suggesting that GW-5638 and raloxifene share a mechanism of action that is different from that of tamoxifen. RESULTS Both GW-5638 and its hydroxylated analog GW-7604 were tested for their ability to bind to ERalpha and ERbeta and their ability to affect transcription of ERalpha and ERbeta at a consensus estrogen response element and an ER/AP-1 response element. The drugs were found to have the same affinity for ERalpha and ERbeta, although they were also found to activate transcription from an AP-1 promoter element more potently with ERbeta than with ERalpha. Derivatives of GW-5638 with alterations at the carboxylic acid still showed increased ERbeta potency compared to ERalpha, but the magnitude of the activation with ERalpha was much higher than with ERbeta. CONCLUSIONS Despite similar binding affinities to isolated ERalpha and ERbeta, GW-5638 and GW-7604 show markedly lower EC(50) values with ERbeta at an AP-1-driven promoter as compared to ERalpha. This suggests that the two compounds produce a more active ER/AP-1 conformation of the ER/AP-1 transcription factor complex when bound to ERbeta than when bound to ERalpha.
Collapse
Affiliation(s)
- R V Weatherman
- Departments of Pharmaceutical Chemistry and Cellular and Molecular Pharmacology, University of California, San Francisco 94143-0446, USA
| | | | | |
Collapse
|
46
|
Muyan M, Yi P, Sathya G, Willmert LJ, Driscoll MD, Hilf R, Bambara RA. Fusion estrogen receptor proteins: toward the development of receptor-based agonists and antagonists. Mol Cell Endocrinol 2001; 182:249-63. [PMID: 11514059 DOI: 10.1016/s0303-7207(01)00493-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Estrogen-induced signaling mediated by estrogen receptors (ERs) is also affected by aberrant ERs that act as constitutively active or dominant negative modulators. Variant ERs can contribute to carcinogenesis and to the loss of estrogen responsiveness, rendering antiestrogen therapy ineffective. Determining target gene response during co-synthesis of different ER species is difficult, because dimers formed in the presence of more than one ER species are a heterogenous population of homo- or heterodimers. We engineered a homofusion ERalpha as a prototype single-chain receptor by genetically conjugating two ER monomers into a covalently fused single-chain protein to obtain a homogeneous population. This permits analysis of symmetrical or asymmetrical mutations that simulate variant homo- and heterodimers. Although a monomer, the homofusion receptor exhibited similar biochemical and functional properties to the dimeric ERalpha. We used activation function-2 (AF2) defective mutants as a model in either one or both receptor domains for a dominant-negative phenotype by suppressing the reporter activity induced by the WT receptor. When co-expressed with ERalpha, the fusion variant deficient in both AF2 functions suppressed the reporter activity effectively induced by ERalpha. These results show the utility of fusion receptors as models for generation of receptor-based agonists and antagonists.
Collapse
Affiliation(s)
- M Muyan
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Jordan V. Oestrogen receptors, growth factors and the control of breast cancer. Breast 2001. [DOI: 10.1016/s0960-9776(16)30006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
48
|
Daxenberger A, Ibarreta D, Meyer HH. Possible health impact of animal oestrogens in food. APMIS 2001. [DOI: 10.1111/j.1600-0463.2001.tb05791.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Signoretti S, Loda M. Estrogen receptor beta in prostate cancer: brake pedal or accelerator? THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:13-6. [PMID: 11438447 PMCID: PMC1850394 DOI: 10.1016/s0002-9440(10)61666-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- S Signoretti
- Department of Adult Oncology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
50
|
Fertuck KC, Kumar S, Sikka HC, Matthews JB, Zacharewski TR. Interaction of PAH-related compounds with the alpha and beta isoforms of the estrogen receptor. Toxicol Lett 2001; 121:167-77. [PMID: 11369471 DOI: 10.1016/s0378-4274(01)00344-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The ability of several 4- and 5-ring polycyclic aromatic hydrocarbons (PAHs), heterocyclic PAHs, and their monohydroxy derivatives to interact with the estrogen receptor (ER) alpha and beta isoforms was examined. Only compounds possessing a hydroxyl group were able to compete with 3H-labeled 17beta-estradiol (E2) for binding to either a glutathione-S-transferase and human ERalpha D, E, and F domain fusion protein (GST-hERalphadef) or to the full-length human ERbeta. Competitive binding was comparable for both isoforms, with IC(50) values ranging from 20 to 300 nM (E2 IC(50) approximately 3 nM). However, several compounds were able to induce reporter gene expression preferentially through mERbeta, using MCF-7 cells transiently transfected with either a Gal4-human ERalphadef or Gal4-mouse ERbetadef construct, as well as a Gal4-regulated reporter. These data extend the number and type of PAH-related compounds capable of interacting with ERalpha and ERbeta, and provides additional evidence that even though some compounds may possess a similar affinity for both ER isoforms, the capacity for transcriptional activation can still be isoform-specific.
Collapse
Affiliation(s)
- K C Fertuck
- Department of Biochemistry and Molecular Biology, National Food Safety and Toxicology Center, Michigan State University, Wilson Road, East Lansing, MI 48824-1319, USA
| | | | | | | | | |
Collapse
|