101
|
MA LI, LIU YUEPING, GENG CUIZHI, QI XIAOWEI, JIANG JUN. Estrogen receptor β inhibits estradiol-induced proliferation and migration of MCF-7 cells through regulation of mitofusin 2. Int J Oncol 2013; 42:1993-2000. [DOI: 10.3892/ijo.2013.1903] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/15/2013] [Indexed: 11/06/2022] Open
|
102
|
17β-estradiol up-regulates miR-155 expression and reduces TP53INP1 expression in MCF-7 breast cancer cells. Mol Cell Biochem 2013; 379:201-11. [PMID: 23568502 DOI: 10.1007/s11010-013-1642-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/28/2013] [Indexed: 02/07/2023]
Abstract
In estrogen responsive breast cancer cells, estradiol (E2) is a key regulator of cell proliferation and survival. MiR-155 has emerged as an "oncomiR", which is the most significantly up-regulated miRNA in breast cancer. Moreover, miR-155 is higher in ERα (+) breast tumors than ERα (-), but no one has examined whether E2 regulates miR-155 expression in MCF-7 cells. In this study, the aim was to explore whether miR-155 involved in E2 regulated expression of estrogen responsive genes. We evaluated miR-155 expression in human breast cancer cells by real-time PCR, finding out miR-155 was overexpressed in MCF-7 cells compared with MDA-MB-231 cells. Treatment with E2 in MCF-7 cells increased miR-155 expression, promoting proliferation and decreasing apoptosis, similarly, transfection of miR-155m to MCF-7 cells gave the similar results. In contrast, inhibited miR-155 expression by transfection with miR-155 inhibitors reduced proliferation and promoted apoptosis of MCF-7 cells. Moreover, TP53INP1 is one of the targets of miR-155. E2 negatively regulated TP53INP1 mRNA expression and the protein expression of TP53INP1, cleaved-caspase-3, -8, -9, and p21, whereas transfection with miR-155 inhibitors increased TP53INP1, cleaved-caspase-3, -8, -9, and p21 protein level. These results demonstrated that E2 promoted breast cancer development and progression possibly through increasing the expression of miR-155, which was overexpressed in MCF-7 cells, contributes to proliferation of MCF-7 cells possibly through down-regulating TP53INP1.
Collapse
|
103
|
Coughlan N, Thillainadesan G, Andrews J, Isovic M, Torchia J. β-Estradiol-dependent activation of the JAK/STAT pathway requires p/CIP and CARM1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1463-75. [PMID: 23434684 DOI: 10.1016/j.bbamcr.2013.02.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 01/15/2013] [Accepted: 02/11/2013] [Indexed: 01/07/2023]
Abstract
The steroid receptor coactivator p/CIP, also known as SRC-3, is an oncogene commonly amplified in breast and ovarian cancers. p/CIP is known to associate with coactivator arginine methyltransferase 1 (CARM1) on select estrogen responsive genes. We have shown, using a ChIP-on-chip approach, that in response to stimulation with 17β-estradiol (E2), the p/CIP/CARM1 complex is recruited to 204 proximal promoters in MCF-7 cells. Many of the complex target genes have been previously implicated in signaling pathways related to oncogenesis. Jak2, a member of the Jak/Stat signaling cascade, is one of the direct E2-dependent targets of the p/CIP/CARM1 complex. Following E2-treatment, histone modifications at the Jak2 promoter are reflective of a transcriptionally permissive gene, and modest changes in RNA and protein expression lead us to suggest that an additional factor(s) may be required for a more notable transcriptional and functional response. Bioinformatic examination of the 204 proximal promoter sequences of p/CIP/CARM1 targets supports the idea that transcription factor crosstalk is likely the favored mechanism of E2-dependent p/CIP/CARM1 complex recruitment. This data may have implications towards understanding the oncogenic role of p/CIP in breast cancer and ultimately allow for the identification of new prognostic indicators and/or viable therapeutic targets.
Collapse
Affiliation(s)
- N Coughlan
- Dept. of Oncology, The University of Western Ontario, Canada
| | | | | | | | | |
Collapse
|
104
|
Norethisterone acetate alters coagulation gene expression in vitro in human cell culture. Thromb Res 2013; 131:72-7. [DOI: 10.1016/j.thromres.2012.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 11/19/2022]
|
105
|
Abstract
Estrogen provides many beneficial effects early in life by regulating normal tissue development and several physiological functions. While estrogen replacement therapy (ERT) in women was expected to reduce the health risks associated with the age-related decline in estrogen levels during menopause, ERT also resulted in increased progression to other types of diseases. Hence, distinguishing the signaling pathways that regulate the beneficial and detrimental effects of estrogen is important for developing interventions that selectively harness the hormone's beneficial effects, while minimizing its side effects. Estrogen can minimize mitochondrial dysfunction, which is thought to contribute to aging phenotypes. Decline in estrogen levels during menopause may lead to progressive mitochondrial dysfunction and may permanently alter cellular response, including that of estrogen (i.e., ERT). This review discusses the interplay between estrogen and mitochondrial function during the aging process and suggests a potential role of mitochondria in influencing the pleiotropic action of estrogen.
Collapse
Affiliation(s)
- Michael C Velarde
- Buck Institute for Research on Aging, Novato, California 94945, USA.
| |
Collapse
|
106
|
Muyan M, Callahan LM, Huang Y, Lee AJ. The ligand-mediated nuclear mobility and interaction with estrogen-responsive elements of estrogen receptors are subtype specific. J Mol Endocrinol 2012; 49:249-66. [PMID: 23014840 PMCID: PMC3674415 DOI: 10.1530/jme-12-0097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
17β-Estradiol (E(2)) plays important roles in functions of many tissues. E(2) effects are mediated by estrogen receptor (ER) α and β. ERs regulate transcriptions through estrogen-responsive element (ERE)-dependent and ERE-independent modes of action. ER binding to ERE constitutes the basis of the ERE-dependent pathway. Direct/indirect ER interactions with transcription complexes define ERE-independent signaling. ERs share functional features. Ligand-bound ERs nevertheless induce distinct transcription profiles. Live cell imaging indicates a dynamic nature of gene expressions by highly mobile ERs. However, the relative contribution of ER mobility at the ERE-independent pathway to the overall kinetics of ER mobility remains undefined. We used fluorescent recovery after a photo-bleaching approach to assess the ligand-mediated mobilities of ERE binding-defective ERs, ER(EBD). The decrease in ERα mobility with E(2) or the selective ER modulator 4-hydroxyl-tamoxifen (4HT) was largely due to the interaction of the receptor with ERE. Thus, ERα bound to E(2) or 4HT mediates transcriptions from the ERE-independent pathway with remarkably fast kinetics that contributes fractionally to the overall motility of the receptor. The antagonist Imperial Chemical Industries 182 780 immobilized ERαs. The mobilities of ERβ and ERβ(EBD) in the presence of ligands were indistinguishable kinetically. Thus, ERβ mobility is independent of the nature of ligands and the mode of interaction with target sites. Chimeric ERs indicated that the carboxyl-termini are critical regions for subtype-specific mobility. Therefore, while ERs are highly mobile molecules interacting with target sites with fast kinetics, an indication of the hit-and-run model of transcription, they differ mechanistically to modulate transcriptions.
Collapse
Affiliation(s)
- Mesut Muyan
- Department of Biochemistry and Biophysics, University of Rochester Medical School, Rochester, New York 14642, USA.
| | | | | | | |
Collapse
|
107
|
Dasgupta C, Chen M, Zhang H, Yang S, Zhang L. Chronic hypoxia during gestation causes epigenetic repression of the estrogen receptor-α gene in ovine uterine arteries via heightened promoter methylation. Hypertension 2012; 60:697-704. [PMID: 22777938 DOI: 10.1161/hypertensionaha.112.198242] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Estrogen receptor-α (ERα) plays a key role in the adaptation of increased uterine blood flow in pregnancy. Chronic hypoxia is a common stress to maternal cardiovascular homeostasis and causes increased risk of preeclampsia. Studies in pregnant sheep demonstrated that hypoxia during gestation downregulated ERα gene expression in uterine arteries. The present study tested the hypothesis that hypoxia causes epigenetic repression of the ERα gene in uterine arteries via heightened promoter methylation. Ovine ERα promoter of 2035 bp spanning from -2000 to +35 of the transcription start site was cloned. No estrogen or hypoxia-inducible factor response elements were found at the promoter. Two transcription factor binding sites, USF(-15) and Sp1(-520), containing CpG dinucleotides were identified, which had significant effects on the promoter activity. The USF element binds transcription factors USF1 and USF2, and the Sp1 element binds Sp1, as well as ERα through Sp1. Deletion of the Sp1 site abrogated 17β-estradiol-induced increase in the promoter activity. In normoxic control sheep, CpG methylation at the Sp1 but not the USF site was significantly decreased in uterine arteries of pregnant as compared with nonpregnant animals. In pregnant sheep exposed to long-term high-altitude hypoxia, CpG methylation at both Sp1 and USF sites in uterine arteries was significantly increased. Methylation inhibited transcription factor binding and the promoter activity. The results provide evidence of hypoxia causing heightened promoter methylation and resultant ERα gene repression in uterine arteries and suggest new insights of molecular mechanisms linking gestational hypoxia to aberrant uteroplacental circulation and increased risk of preeclampsia.
Collapse
Affiliation(s)
- Chiranjib Dasgupta
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | |
Collapse
|
108
|
Bartella V, Rizza P, Barone I, Zito D, Giordano F, Giordano C, Catalano S, Mauro L, Sisci D, Panno ML, Fuqua SAW, Andò S. Estrogen receptor beta binds Sp1 and recruits a corepressor complex to the estrogen receptor alpha gene promoter. Breast Cancer Res Treat 2012; 134:569-81. [PMID: 22622808 DOI: 10.1007/s10549-012-2090-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 05/03/2012] [Indexed: 02/06/2023]
Abstract
Human estrogen receptors alpha and beta are crucially involved in the regulation of mammary growth and development. Normal breast tissues display a relative higher expression of ER beta than ER alpha, which drastically changes during breast tumorogenesis. Thus, it is reasonable to suggest that a dysregulation of the two estrogen receptor subtypes may induce breast cancer development. However, the molecular mechanisms underlying the potential opposing roles played by the two estrogen receptors on tumor cell growth remain to be elucidated. In the present study, we have demonstrated that ER beta overexpression in breast cancer cells decreases cell proliferation and down-regulates ER alpha mRNA and protein content, along with a concomitant repression of estrogen-regulated genes. Transient transfection experiments, using a vector containing the human ER alpha promoter region, showed that elevated levels of ER beta down-regulated basal ER alpha promoter activity. Furthermore, site-directed mutagenesis and deletion analysis revealed that the proximal GC-rich motifs at -223 and -214 are critical for the ER beta-induced ER alpha down-regulation in breast cancer cells. This occurred through ER beta-Sp1 protein-protein interactions within the ER alpha promoter region and the recruitment of a corepressor complex containing the nuclear receptor corepressor NCoR, accompanied by hypoacetylation of histone H4 and displacement of RNA-polymerase II. Silencing of NCoR gene expression by RNA interference reversed the down-regulatory effects of ER beta on ER alpha gene expression and cell proliferation. Our results provide evidence for a novel mechanism by which overexpression of ER beta through NCoR is able to down regulate ER alpha gene expression, thus blocking ER alpha's driving role on breast cancer cell growth.
Collapse
Affiliation(s)
- V Bartella
- Department of Pharmaco-Biology, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, CS, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Glait-Santar C, Benayahu D. Regulation of SVEP1 gene expression by 17β-estradiol and TNFα in pre-osteoblastic and mammary adenocarcinoma cells. J Steroid Biochem Mol Biol 2012; 130:36-44. [PMID: 22265959 DOI: 10.1016/j.jsbmb.2011.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 12/21/2011] [Accepted: 12/22/2011] [Indexed: 10/14/2022]
Abstract
Breast cancer is one of several tumors, including prostate, thyroid and kidney, which display a remarkable predilection for metastasis to bone. The preference to metastasize to bone by tumor cells relies on specific interactions among tumor cells, bone marrow microenvironment and bone cells. Osteomimicry is postulated to enable the survival of tumor cells in the bone tissue. Using gene profiling array and RT-PCR we demonstrated the message expression of few bone matrix proteins in mammary adenocarcinoma cells as well as that of cell adhesion molecules (CAMs). A CAM molecule, named SVEP1, was previously shown to be expressed in osteoblastic cells both in vivo and in vitro mediating cell adhesion in the bone-marrow niches. Both estradiol (17βE(2)) and TNFα regulate the expression of adhesion molecules and act in bone-cancer-crosstalk. We focused on differential regulation of SVEP1 gene comparing pre-osteoblastic MBA-15 and mammary adenocarcinoma DA3 cells. 17βE(2) and TNFα activated SVEP1 promoter, increased its message and protein levels in both cell types. Using chromatin immunoprecipitation assay, we quantified SVEP1 promoter occupancy by transcription factors; TFIIB, ERα, NF-κB, Sp1 and their binding was also regulated by both factors. By comparing pre-osteoblastic with mammary adenocarcinoma cells, the study expands our understanding of SVEP1 gene expression regulation and it sheds light on its involvement in bone-cancer-microenvironment interactions.
Collapse
Affiliation(s)
- C Glait-Santar
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.
| | | |
Collapse
|
110
|
Cunningham MA, Naga OS, Eudaly JG, Scott JL, Gilkeson GS. Estrogen receptor alpha modulates Toll-like receptor signaling in murine lupus. Clin Immunol 2012; 144:1-12. [PMID: 22659029 DOI: 10.1016/j.clim.2012.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 01/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is a disease that disproportionately affects females. Despite significant research effort, the mechanisms underlying the female predominance in this disease are largely unknown. Previously, we showed that estrogen receptor alpha knockout (ERαKO) lupus prone female mice had significantly less pathologic renal disease and proteinuria, and significantly prolonged survival. Since autoantibody levels and number and percentage of B/T cells were not significantly impacted by ERα genotype, we hypothesized that the primary benefit of ERα deficiency in lupus nephritis was via modulation of the innate immune response. Using BMDCs and spleen cells/B cells from female wild-type or ERαKO mice, we found that ERαKO-derived cells have a significantly reduced inflammatory response after stimulation with TLR agonists. Our results indicate that the inflammatory response to TLR ligands is significantly impacted by the presence of ERα despite the absence of estradiol, and may partially explain the protective effect of ERα deficiency in lupus-prone animals.
Collapse
Affiliation(s)
- Melissa A Cunningham
- Medical University of South Carolina, Division of Rheumatology and Immunology and Ralph H. Johnson Veterans Affairs Hospital, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
111
|
Wang S, Zhu X, Cong B, You X, Wang Y, Wang W, Ni X. Estrogenic action on arterial smooth muscle: permissive for maintenance of CRHR2 expression. Endocrinology 2012; 153:1915-24. [PMID: 22315451 DOI: 10.1210/en.2011-1939] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Urocortin (Ucn), a member of CRH family, has been implicated to be one of the endogenous regulators in the cardiovascular system and exerts its effects locally via an autocrine/paracrine fashion. Previous studies have shown the gender difference in CRH-induced vasodilation in human skin, which is related to the concentration of estrogens during the menstrual cycle. The aim of this study was to investigate whether estrogens modulate Ucn/CRH receptor type 2 (CRHR2) expression in vascular smooth muscle, thereby leading to vasodilation. We performed sham operation or bilateral ovariectomy (OVX) on female Sprague Dawley rats. OVX rats were sc administered 17β-estradiol (E₂) at a dose of 30 μg/kg·d or with placebo for 12 wk. Primary smooth muscle cells of aorta were used for the in vitro study. It was found that the Ucn-induced vasodilation and CRHR2 expression were decreased in OVX rats and restored by E₂ replacement treatment for 12 wk. E₂ increased the expression of CRHR2 in cultured smooth muscle cells, which was blocked by estrogen receptor-β antagonist. Ucn significantly suppressed the phenylephrine-induced phospholipase Cβ3 activation, inositol 1,4,5-trisphosphate (IP₃) production, and intracellular Ca²⁺ elevation. Ucn stimulated the expression of active GTP-bound Gαs protein and cAMP production. The suppressive effects of Ucn on phenylephrine-induced IP₃ production and intracellular Ca²⁺ elevation were blocked by the inhibitors of adenylate cyclase and protein kinase A. Our results demonstrate that estrogen maintains the expression of CRHR2 in aorta smooth muscle, thereby enhancing vasodilator actions of Ucn. Ucn exerts its vasorelaxant effects via Gαs-cAMP-protein kinase A signaling, leading to down-regulation of the phospholipase Cβ-IP₃-Ca²⁺ signaling pathway.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Calcium Signaling/physiology
- Cells, Cultured
- Estradiol/pharmacology
- Estrogen Receptor beta/antagonists & inhibitors
- Estrogens/pharmacology
- Female
- In Vitro Techniques
- Models, Animal
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Ovariectomy
- Rats
- Rats, Sprague-Dawley
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Signal Transduction/physiology
- Urocortins/metabolism
- Vasodilation/drug effects
- Vasodilation/physiology
Collapse
Affiliation(s)
- Shan Wang
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
112
|
Yu X, Zhang X, Dhakal IB, Beggs M, Kadlubar S, Luo D. Induction of cell proliferation and survival genes by estradiol-repressed microRNAs in breast cancer cells. BMC Cancer 2012; 12:29. [PMID: 22260523 PMCID: PMC3274428 DOI: 10.1186/1471-2407-12-29] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/20/2012] [Indexed: 12/28/2022] Open
Abstract
Background In estrogen responsive MCF-7 cells, estradiol (E2) binding to ERα leads to transcriptional regulation of genes involved in the control of cell proliferation and survival. MicroRNAs (miRNAs) have emerged as key post-transcriptional regulators of gene expression. The aim of this study was to explore whether miRNAs were involved in hormonally regulated expression of estrogen responsive genes. Methods Western blot and QPCR were used to determine the expression of estrogen responsive genes and miRNAs respectively. Target gene expression regulated by miRNAs was validated by luciferase reporter assays and transfection of miRNA mimics or inhibitors. Cell proliferation was evaluated by MTS assay. Results E2 significantly induced bcl-2, cyclin D1 and survivin expression by suppressing the levels of a panel of miRNAs (miR-16, miR-143, miR-203) in MCF-7 cells. MiRNA transfection and luciferase assay confirmed that bcl-2 was regulated by miR-16 and miR-143, cyclinD1 was modulated by miR-16. Importantly, survivin was found to be targeted by miR-16, miR-143, miR-203. The regulatory effect of E2 can be either abrogated by anti-estrogen ICI 182, 780 and raloxifene pretreatment, or impaired by ERα siRNA, indicating the regulation is dependent on ERα. In order to investigate the functional significance of these miRNAs in estrogen responsive cells, miRNAs mimics were transfected into MCF-7 cells. It revealed that overexpression of these miRNAs significantly inhibited E2-induced cell proliferation. Further study of the expression of the miRNAs indicated that miR-16, miR-143 and miR-203 were highly expressed in triple positive breast cancer tissues, suggesting a potential tumor suppressing effect of these miRNAs in ER positive breast cancer. Conclusions These results demonstrate that E2 induces bcl-2, cyclin D1 and survivin by orchestrating the coordinate downregulation of a panel of miRNAs. In turn, the miRNAs manifest growth suppressive effects and control cell proliferation in response to E2. This sheds a new insight into the integral post-transcriptional regulation of cell proliferation and survival genes by miRNAs, a potential therapeutic option for breast cancer.
Collapse
Affiliation(s)
- Xinfeng Yu
- Department of Pharmacology, School of Chemical Biology & Pharmaceutical Sciences, Capital Medical University, 100069 Beijing, China.
| | | | | | | | | | | |
Collapse
|
113
|
Aquino NB, Sevigny MB, Sabangan J, Louie MC. The role of cadmium and nickel in estrogen receptor signaling and breast cancer: metalloestrogens or not? JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2012; 30:189-224. [PMID: 22970719 PMCID: PMC3476837 DOI: 10.1080/10590501.2012.705159] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
During the past half-century, incidences of breast cancer have increased globally. Various factors--genetic and environmental--have been implicated in the initiation and progression of this disease. One potential environmental risk factor that has not received a lot of attention is the exposure to heavy metals. While several mechanisms have been put forth describing how high concentrations of heavy metals play a role in carcinogenesis, it is unclear whether chronic, low-level exposure to certain heavy metals (i.e., cadmium and nickel) can directly result in the development and progression of cancer. Cadmium and nickel have been hypothesized to play a role in breast cancer development by acting as metalloestrogens--metals that bind to estrogen receptors and mimic the actions of estrogen. Since the lifetime exposure to estrogen is a well-established risk factor for breast cancer, anything that mimics its activity will likely contribute to the etiology of the disease. However, heavy metals, depending on their concentration, are capable of binding to a variety of proteins and may exert their toxicities by disrupting multiple cellular functions, complicating the analysis of whether heavy metal-induced carcinogenesis is mediated by the estrogen receptor. The purpose of this review is to discuss the various epidemiological, in vivo, and in vitro studies that show a link between the heavy metals, cadmium and nickel, and breast cancer development. We will particularly focus on the studies that test whether these two metals act as metalloestrogens in order to assess the strength of the data supporting this hypothesis.
Collapse
Affiliation(s)
- Natalie B. Aquino
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael CA 94901
| | - Mary B. Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael CA 94901
| | - Jackielyn Sabangan
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael CA 94901
| | - Maggie C. Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael CA 94901
| |
Collapse
|
114
|
Zhao R. Immune regulation of osteoclast function in postmenopausal osteoporosis: a critical interdisciplinary perspective. Int J Med Sci 2012; 9:825-32. [PMID: 23136547 PMCID: PMC3491443 DOI: 10.7150/ijms.5180] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 10/15/2012] [Indexed: 01/11/2023] Open
Abstract
Extensive studies on cross talk between immune and skeletal systems in autoimmune diseases give rise to a new discipline of 'osteoimmunology', which explores the molecular regulation of osteoclasts by immune system. Postmenopausal osteoporosis is recognized as a cytokine driven disease, but the mechanism that how estrogen deficiency interplaying with cytokines to stimulate bone loss remains to be elucidated. Although the effect of individual cytokines on osteoclast formation is well characterized, the major challenge is to fit a multitude of redundant pathways and cytokines into a systemic model of postmenopausal osteoporosis. This review presents current findings and hypothesis to explain estrogen deficiency-stimulated bone loss in a critical interdisciplinary perspective. To better understand the interaction between osteoclasts and immune system in postmenopausal osteoporosis, many of the lessons have been explored in animal models.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Zhejiang, Jinhua, China.
| |
Collapse
|
115
|
Ahn SE, Jeong W, Kim JH, Lim W, Kim J, Bazer FW, Han JY, Song G. ERBB receptor feedback inhibitor 1: identification and regulation by estrogen in chickens. Gen Comp Endocrinol 2012; 175:194-205. [PMID: 22137914 DOI: 10.1016/j.ygcen.2011.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 10/26/2011] [Accepted: 11/11/2011] [Indexed: 11/16/2022]
Abstract
The ERBB receptor feedback inhibitor 1 (ERRFI1) is a scaffolding adaptor protein, that plays a pivotal role in the epidermal growth factor receptor (EGFR) cell signaling cascade as a negative regulator affecting many important physiological processes. It was recently reported that ERRFI1 is a critical regulator of the response of the endometrium to estrogen regulation of tissue homeostasis in mice. But, very little is known about ERRF11 and hormonal regulation of the ERRFI1 gene in chickens. Therefore, in the present study, ERRFI1 gene was cloned and its differential expression profile analyzed at different embryonic stages, in various adult organs, and in oviducts from estrogen-treated chickens. Chicken ERRFI1 has an open-reading frame of 2848 nucleotides that encode for a protein of 465 amino acids that has considerable homology to mammalian ERRFI1 proteins (>62% identity). Importantly, ERRFI1 mRNA is abundantly distributed in various organs from chickens. We then determined that DES (diethylstilbestrol, a synthetic nonsteroidal estrogen) induced ERRFI1 mRNA and protein predominantly in luminal and glandular epithelial cells of the oviduct. Further, we determined whether microRNAs, specifically miR-200b, miR-429 and miR-1639, influence ERRFI1 expression via its 3'UTR and found that it does not directly target the 3'UTR of ERRFI1 mRNA. Therefore, it is unlikely that post-transcriptional regulation influences ERRFI1 expression in the chicken oviduct. In conclusion, our results indicate that ERRFI1 is a novel estrogen-stimulated gene expressed in epithelial cells of the chicken oviduct that likely plays an important role in oviduct growth and differentiation during early development of the chicken.
Collapse
Affiliation(s)
- Suzie E Ahn
- WCU Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-921, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Sreeja S, Santhosh Kumar TR, Lakshmi BS, Sreeja S. Pomegranate extract demonstrate a selective estrogen receptor modulator profile in human tumor cell lines and in vivo models of estrogen deprivation. J Nutr Biochem 2011; 23:725-32. [PMID: 21839626 DOI: 10.1016/j.jnutbio.2011.03.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 03/17/2011] [Indexed: 02/03/2023]
Abstract
Selective estrogen receptor modulators (SERMs) are estrogen receptor (ER) ligands exhibiting tissue-specific agonistic or antagonistic biocharacter and are used in the hormonal therapy for estrogen-dependent breast cancers. Pomegranate fruit has been shown to exert antiproliferative effects on human breast cancer cells in vitro. In this study, we investigated the tissue-specific estrogenic/antiestrogenic activity of methanol extract of pericarp of pomegranate (PME). PME was evaluated for antiproliferative activity at 20-320 μg/ml on human breast (MCF-7, MDA MB-231) endometrial (HEC-1A), cervical (SiHa, HeLa), ovarian (SKOV3) carcinoma and normal breast fibroblast (MCF-10A) cells. Competitive radioactive binding studies were carried out to ascertain whether PME interacts with ER. The reporter gene assay measured the estrogenic/antiestrogenic activity of PME in MCF-7 and MDA MB-231 cells transiently transfected with plasmids coding estrogen response elements with a reporter gene (pG5-ERE-luc) and wild-type ERα (hEG0-ER). PME inhibited the binding of [³H] estradiol to ER and suppressed the growth and proliferation of ER-positive breast cancer cells. PME binds ER and down-regulated the transcription of estrogen-responsive reporter gene transfected into breast cancer cells. The expressions of selected estrogen-responsive genes were down-regulated by PME. Unlike 17β-estradiol [1 mg/kg body weight (BW)] and tamoxifen (10 mg/kg BW), PME (50 and 100 mg/kg BW) did not increase the uterine weight and proliferation in ovariectomized mice and its cardioprotective effects were comparable to that of 17β-estradiol. In conclusion, our findings suggest that PME displays a SERM profile and may have the potential for prevention of estrogen-dependent breast cancers with beneficial effects in other hormone-dependent tissues.
Collapse
Affiliation(s)
- Sreekumar Sreeja
- Integrated Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O., Thiruvananthapuram, Kerala 695014, India.
| | | | | | | |
Collapse
|
117
|
Acconcia F, Marino M. The Effects of 17β-estradiol in Cancer are Mediated by Estrogen Receptor Signaling at the Plasma Membrane. Front Physiol 2011; 2:30. [PMID: 21747767 PMCID: PMC3129035 DOI: 10.3389/fphys.2011.00030] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/17/2011] [Indexed: 12/16/2022] Open
Abstract
Two different isoforms of the estrogen receptors (i.e., ERα and ERβ) mediate pleiotropic 17β-estradiol (E2)-induced cellular effects. The ERs are principally localized in the nucleus where they act by globally modifying the expression of the E2-target genes. The premise that E2 effects are exclusively mediated through the nuclear localized ERs has been rendered obsolete by research over the last 15 years demonstrating that ERα and ERβ proteins are also localized at the plasma membranes and in other extra-nuclear organelles. The E2 modulation of cancer cell proliferation represents a good example of the impact of membrane-initiated signals on E2 effects. In fact, E2 via ERα elicits rapid signals driving cancer cells to proliferation (e.g., in breast cancer cells), while E2-induced ERβ rapid signaling inhibits proliferation (e.g., in colon cancer cells). In this review we provide with an overview of the complex system of E2-induced signal transduction pathways, their impact on E2-induced cancer cell proliferation, and the participation of E2-induced membrane-initiated signals in tumor environment.
Collapse
Affiliation(s)
- Filippo Acconcia
- Cell Physiology Laboratory, Department of Biology, University Roma Tre Rome, Italy
| | | |
Collapse
|
118
|
Burns KA, Li Y, Arao Y, Petrovich RM, Korach KS. Selective mutations in estrogen receptor alpha D-domain alters nuclear translocation and non-estrogen response element gene regulatory mechanisms. J Biol Chem 2011; 286:12640-9. [PMID: 21285458 PMCID: PMC3069464 DOI: 10.1074/jbc.m110.187773] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 01/28/2011] [Indexed: 01/05/2023] Open
Abstract
The three main mechanisms of ERα action are: 1) nuclear, genomic, direct DNA binding, 2) nuclear, genomic, "tethered"-mediated, protein-protein interactions, and 3) non-nuclear, non-genomic, rapid action responses. Reports suggest the D-domain or hinge region of ERα plays an important role in mechanisms 1 and 2 above. Studies demonstrating the functionality of the ERα hinge region have resected the full D-domain; therefore, site directed mutations were made to attribute precise sequence functionality to this domain. This study focuses on the characterization and properties of three novel site directed ERα- D-domain mutants. The Hinge 1 (H1) ERα mutant has disrupted nuclear localization, can no longer perform tethered mediated responses and has lost interaction with c-Jun, but retains estrogen response element (ERE)-mediated functions as demonstrated by confocal microscopy, reporter assays, endogenous gene expression and co-immunoprecipitation. The H2 ERα mutant is non-nuclear, but translocates to the nucleus with estradiol (E2) treatment and maintains ERE-mediated functionality. The H2+NES ERα mutant does not maintain nuclear translocation with hormone binding, no longer activates ERE-target genes, functions in ERE- or tethered-mediated luciferase assays, but does retain the non-genomic, non-nuclear, rapid action response. These studies reveal the sequence(s) in the ERα hinge region that are involved in tethered-mediated actions as well as nuclear localization and attribute important functionality to this region of the receptor. In addition, the properties of these ERα mutants will allow future studies to further dissect and characterize the three main ERα mechanisms of action and determine the mechanistic role each action has in estrogen hormone regulation.
Collapse
Affiliation(s)
- Katherine A. Burns
- From Receptor Biology, Laboratory of Reproductive and Developmental Toxicology and
| | - Yin Li
- From Receptor Biology, Laboratory of Reproductive and Developmental Toxicology and
| | - Yukitomo Arao
- From Receptor Biology, Laboratory of Reproductive and Developmental Toxicology and
| | - Robert M. Petrovich
- Protein Expression Core Facility, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Kenneth S. Korach
- From Receptor Biology, Laboratory of Reproductive and Developmental Toxicology and
| |
Collapse
|
119
|
Fujiwara Y, Kiyota N, Tsurushima K, Yoshitomi M, Mera K, Sakashita N, Takeya M, Ikeda T, Araki T, Nohara T, Nagai R. Natural compounds containing a catechol group enhance the formation of Nε-(carboxymethyl)lysine of the Maillard reaction. Free Radic Biol Med 2011; 50:883-91. [PMID: 21195168 DOI: 10.1016/j.freeradbiomed.2010.12.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 12/16/2010] [Accepted: 12/22/2010] [Indexed: 02/07/2023]
Abstract
Inhibition of advanced glycation end-product (AGE) formation is a potential strategy for the prevention of clinical diabetes complications. Screening for new AGE inhibitors revealed several natural compounds that inhibited the formation of N(ε)-(carboxymethyl)lysine (CML), a major antigenic AGE structure, whereas natural compounds containing a catechol group, such as gallic acid and epicatechin, significantly enhanced CML formation. A similar enhancing effect was also observed by culturing THP-1 macrophages in the presence of catechol compounds. Although 4-methylcatechol significantly enhanced CML formation from glycated HSA (gHSA), a model for Amadori proteins, analogues of catechol such as 5-methylresorcinol and methylhydroquinone showed no enhancing effect. Even though 1mM 4-methylcatechol, epicatechin, and gallic acid significantly enhanced CML formation from gHSA, it was significantly inhibited by decreasing their concentration. The enhancing effect of 1mM catechol compounds was inhibited in the presence of the glutathione peroxidase system, thus demonstrating that hydrogen peroxide generated from catechol compounds plays an important role in the enhancement of CML formation. Furthermore, administration of 500mg/kg/day epicatechin to STZ-induced diabetic mice for 45days enhanced CML accumulation at the surface area of gastric epithelial cells in the stomach. This study provides the first evidence that high amounts of catechol-containing structures enhance oxidative stress, thus leading to enhanced CML formation, and this phenomenon may explain the paradoxical effect that some flavonoids have on redox status.
Collapse
Affiliation(s)
- Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Abstract
Due to the female predominance of autoimmune diseases, the role of gender and sex hormones in the immune system is of long-term interest. Estrogen's primary effects are mediated via estrogen receptors alpha and beta (ER α/β) that are expressed on most immune cells. ERs are nuclear hormone receptors that can either directly bind to estrogen response elements in gene promoters or serve as cofactors with other transcription factors (i.e., NFkB/AP1). Cytoplasmic ER and membrane associated ER impact specific kinase signaling pathways. ERs have prominent effects on immune function in both the innate and adaptive immune responses. Genetic deficiency of ERα in murine models of lupus resulted in significantly decreased disease and prolonged survival, while ERβ deficiency had minimal to no effect in autoimmune models. The protective effect of ERα in lupus is multifactoral. In arthritis models, ERα agonists appears to mediate a protective effect. The modulation of ERα function appears to be a potential target for therapy in autoimmunity.
Collapse
Affiliation(s)
- Melissa Cunningham
- Medical Research Service, Ralph H. Johnson VAMC and the Department of Medicine, Division of Rheumatology, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 912, Charleston, SC 29425, USA
| | | |
Collapse
|
121
|
MDM2 SNP309 is associated with endometrial cancer susceptibility: a meta-analysis. Hum Cell 2011; 24:57-64. [PMID: 21547352 DOI: 10.1007/s13577-011-0013-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 11/24/2010] [Indexed: 10/18/2022]
Abstract
Epidemiological studies have investigated the association between MDM2 promoter SNP 309 (T/G) and endometrial cancer susceptibility. However, the results are still controversial. To obtain a more precise estimate of the relationship, we conducted a meta-analysis of 1,001 cases and 1,889 controls from 6 published case-control studies (one of five articles contains two studies) to estimate the effect of SNP309 on endometrial cancer risk. The strength of association between MDM2 SNP309 and endometrial cancer susceptibility was assessed by calculating pooled odds ratios (ORs) with 95% confidence intervals (CIs). When all the eligible studies were pooled in the meta-analysis, we found that elevated endometrial cancer risk was significantly associated with GG variant genotype, however, heterozygous genotype TG seemed to be only a minor modifier on endometrial cancer risk (for GG vs. TT, OR = 1.54, 95% CI = 1.21-1.95, P = 0.0004; for TG vs. TT, OR = 0.96, 95% CI = 0.81-1.14, P = 0.66; for dominant model, OR = 1.09, 95% CI = 0.93-1.29, P = 0.29; for recessive model, OR = 1.65, 95% CI = 1.33-2.04, P < 0.00001). Overall, the meta-analysis suggested that the GG genotype of MDM2 SNP309 was significantly associated with the increased endometrial cancer risk.
Collapse
|
122
|
Shehu A, Albarracin C, Devi YS, Luther K, Halperin J, Le J, Mao J, Duan RW, Frasor J, Gibori G. The stimulation of HSD17B7 expression by estradiol provides a powerful feed-forward mechanism for estradiol biosynthesis in breast cancer cells. Mol Endocrinol 2011; 25:754-66. [PMID: 21372145 DOI: 10.1210/me.2010-0261] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Our laboratory has previously cloned and purified an ovarian protein found to be a novel 17β-hydroxysteroid dehydrogenase type 7 enzyme (HSD17B7) (formerly prolactin receptor-associated protein) that converts the weak estrogen, estrone, to the highly potent estradiol. The regulation of this enzyme has not yet been explored. In this report, we show high expression of HSD17B7 in human ductal carcinoma and breast cancer cell lines and present evidence for a strong up-regulation of this enzyme by estradiol at the level of mRNA, protein expression, and promoter activity in MCF-7 cells. The effect of estradiol is mediated by estrogen receptor (ER)α, whereas ERβ prevents this stimulation. ER antagonists, ICI 182,780 and 4-hydroxytamoxifen, prevent estradiol-induced stimulation of the endogenously expressed HSD17B7, suggesting that these inhibitors not only block estradiol action but also its production. We have identified a -185-bp region of the hsd17b7 promoter that is highly conserved among rat, mouse, and human and confers regulation by estradiol in MCF-7 cells. This region is devoid of a classical estradiol-response element but contains a nuclear factor 1 (NF1) site that is essential for estradiol action. We found that estradiol stimulates the recruitment and DNA binding of NF1 to this region of the hsd17b7 promoter. Furthermore, knockdown of NF1 family members, NF1B, NF1A, and NF1X, completely prevents induction of this gene by estradiol. In summary, our findings demonstrate that estradiol stimulates HSD17B7 transcriptional activity in breast cancer cells through a novel mechanism requiring NF1 and strongly suggest a positive feedback mechanism to increase local estradiol synthesis causing growth of estrogen-dependent breast cancers.
Collapse
Affiliation(s)
- Aurora Shehu
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Huang Y, Li X, Muyan M. Estrogen receptors similarly mediate the effects of 17β-estradiol on cellular responses but differ in their potencies. Endocrine 2011; 39:48-61. [PMID: 21069581 PMCID: PMC3683410 DOI: 10.1007/s12020-010-9411-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 10/04/2010] [Indexed: 02/06/2023]
Abstract
17β-estradiol (E2), as the main circulating estrogen hormone, plays critical roles in the physiology and pathophysiology of various tissues. The E2 information is primarily conveyed by the transcription factors, estrogen receptors (ERs) α and β. ERs share similar structural and functional features. Experimental studies indicate that upon binding to E2, ERs directly or indirectly interact with DNA and regulate gene expressions with ERα being more potent transregulator than ERβ. However, studies also showed that ERβ induces alterations in phenotypic features of cancer cell lines independent of E2. These observations suggested that the manner in which the unliganded ERβ induces phenotypic alterations in cancer cell models differs from that of ERα. Studies demonstrated that while requiring E2 for function at low levels of synthesis, the unliganded ERα at augmented concentrations modulates gene expressions and cellular growth. We, therefore, anticipated that heightened levels of ERβ synthesis could similarly circumvent the dependency on E2 leading to gene transcriptions and cellular proliferation. To test this prediction, we used adenovirus-infected cancer cell lines in which ERs were shown to induce genomic and cellular responses. We found that while ERβ at low levels of synthesis was dependent upon E2 for function, the receptor at high levels regulated gene expression and cellular proliferation independent of E2. We then addressed whether ERs at comparable levels that require E2 for function differentially alter gene expressions and cellular responses. We found that ERs mediate the effects of E2 on gene expression, cellular proliferation, apoptosis, and motility with an overlapping pattern. However, ERα was more potent regulator than ERβ in inducing cellular responses. Our results suggest that differences in potencies to regulate the expression of genes are a critical feature of the ER subtypes in mediating E2 signaling in cancer cell lines.
Collapse
Affiliation(s)
- Yanfang Huang
- Department of Biochemistry and Biophysics, University of Rochester Medical School, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
124
|
Habauzit D, Flouriot G, Pakdel F, Saligaut C. Effects of estrogens and endocrine-disrupting chemicals on cell differentiation-survival-proliferation in brain: contributions of neuronal cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:300-327. [PMID: 21790314 DOI: 10.1080/10937404.2011.578554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Estrogens and estrogen receptors (ER) are key actors in the control of differentiation and survival and act on extrareproductive tissues such as brain. Thus, estrogens may display neuritogenic effects during development and neuroprotective effects in the pathophysiological context of brain ischemia and neurodegenerative pathologies like Alzheimer's disease or Parkinson's disease. Some of these effects require classical transcriptional "genomic" mechanisms through ER, whereas other effects appear to rely clearly on "membrane-initiated mechanisms" through cytoplasmic signal transduction pathways. Disturbances of these mechanisms by endocrine-disrupting chemicals (EDC) may exert adverse effects on brain. Some EDC may act via ER-independent mechanisms but might cross-react with endogenous estrogen. Other EDC may act through ER-dependent mechanisms and display agonistic/antagonistic estrogenic properties. Because of these potential effects of EDC, it is necessary to establish sensitive cell-based assays to determine EDC effects on brain. In the present review, some effects of estrogens and EDC are described with focus on ER-mediated effects in neuronal cells. Particular attention is given to PC12 cells, an interesting model to study the mechanisms underlying ER-mediated differentiating and neuroprotective effects of estrogens.
Collapse
Affiliation(s)
- Denis Habauzit
- UMR CNRS 6026 (Interactions Cellulaires et Moléculaires, Equipe RED), Université de Rennes 1, Rennes, France
| | | | | | | |
Collapse
|
125
|
Bratton MR, Duong BN, Elliott S, Weldon CB, Beckman BS, McLachlan JA, Burow ME. Regulation of ERalpha-mediated transcription of Bcl-2 by PI3K-AKT crosstalk: implications for breast cancer cell survival. Int J Oncol 2010; 37:541-50. [PMID: 20664923 DOI: 10.3892/ijo_00000703] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Both estrogen, through the estrogen receptor (ER), and growth factors, through the phosphatidylinositol-3-kinase (PI3K)-AKT pathway, have been shown to independently promote cell survival. Here, we investigated the role of ER/PI3K-AKT crosstalk in the regulation of cell survival in MCF-7 breast carcinoma cells. The ER inhibitor ICI 182,780 was used to determine the requirement of the ER for estrogen in the suppression of tumor necrosis factor-alpha (TNFalpha) induced apoptosis. Gene reporter assays and Western blot analyses were used to determine the involvement of the pro-survival factor Bcl-2 and the coactivator GRIP1 in this survival crosstalk. We demonstrated that an intact ER signaling pathway was required for estrogen to suppress apoptosis induced by TNFalpha. Our gene reporter assays revealed that ERalpha, not ERbeta, was targeted by AKT, resulting in transcriptional potentiation of the full-length Bcl-2 promoter, ultimately leading to increased Bcl-2 protein levels. AKT targeted both activation function (AF) domains of the ERalpha for maximal induction of Bcl-2 reporter activity, although the AF-II domain was predominately targeted. In addition, AKT also caused an upregulation of GRIP1 protein levels. Finally, AKT and GRIP1 cooperated to increase Bcl-2 protein expression to a greater level than either factor alone. Collectively, our study suggests a role for ER/PI3K-AKT crosstalk in cell survival and documents the ability of AKT to regulate Bcl-2 expression via differential activation of ERalpha and ERbeta as well as regulation of GRIP1.
Collapse
Affiliation(s)
- Melyssa R Bratton
- Tulane University School of Medicine, Department of Pharmacology, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | | | | | | | | | | | | |
Collapse
|
126
|
Kretzer NM, Cherian MT, Mao C, Aninye IO, Reynolds PD, Schiff R, Hergenrother PJ, Nordeen SK, Wilson EM, Shapiro DJ. A noncompetitive small molecule inhibitor of estrogen-regulated gene expression and breast cancer cell growth that enhances proteasome-dependent degradation of estrogen receptor {alpha}. J Biol Chem 2010; 285:41863-73. [PMID: 21041310 DOI: 10.1074/jbc.m110.183723] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mechanisms responsible for 17β-estradiol (E(2))-stimulated breast cancer growth and development of resistance to tamoxifen and other estrogen receptor α (ERα) antagonists are not fully understood. We describe a new tool for dissecting ERα action in breast cancer, p-fluoro-4-(1,2,3,6,-tetrahydro-1,3-dimethyl-2-oxo-6-thionpurin-8-ylthio) (TPSF), a potent small-molecule inhibitor of estrogen receptor α that does not compete with estrogen for binding to ERα. TPSF noncompetitively inhibits estrogen-dependent ERα-mediated gene expression with little inhibition of transcriptional activity by NF-κB or the androgen or glucocorticoid receptor. TPSF inhibits E(2)-ERα-mediated induction of the proteinase inhibitor 9 gene, which is activated by ERα binding to estrogen response element DNA, and the cyclin D1 gene, which is induced by tethering ERα to other DNA-bound proteins. TPSF inhibits anchorage-dependent and anchorage-independent E(2)-ERα-stimulated growth of MCF-7 cells but does not inhibit growth of ER-negative MDA-MB-231 breast cancer cells. TPSF also inhibits ERα-dependent growth in three cellular models for tamoxifen resistance; that is, 4-hydroxytamoxifen-stimulated MCF7ERαHA cells that overexpress ERα, fully tamoxifen-resistant BT474 cells that have amplified HER-2 and AIB1, and partially tamoxifen-resistant ZR-75 cells. TPSF reduces ERα protein levels in MCF-7 cells and several other cell lines without altering ERα mRNA levels. The proteasome inhibitor MG132 abolished down-regulation of ERα by TPSF. Thus, TPSF affects receptor levels at least in part due to its ability to enhance proteasome-dependent degradation of ERα. TPSF represents a novel class of ER inhibitor with significant clinical potential.
Collapse
Affiliation(s)
- Nicole M Kretzer
- Department of Biochemistry, University of Illinois, Urbana, Illinois 61801-3602, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Gomes GRO, Yasuhara F, Siu ER, Fernandes SAF, Avellar MCW, Lazari MFM, Porto CS. In vivo treatments with fulvestrant and anastrozole differentially affect gene expression in the rat efferent ductules. Biol Reprod 2010; 84:52-61. [PMID: 20826728 DOI: 10.1095/biolreprod.110.085340] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Estrogen plays a key role in maintaining the morphology and function of the efferent ductules. We previously demonstrated that the antiestrogen fulvestrant markedly affected gene expression in the rat efferent ductules. The mechanism of fulvestrant action to modulate gene expression may involve not only the blockade of ESR1 and ESR2 estrogen receptors, but also the activation of ESR1 and ESR2 when the receptors are tethered to AP-1 or SP1 transcription factors, or the activation of the G protein-coupled estrogen receptor 1. We therefore compared the effects of two strategies to interfere with estrogen action in the rat efferent ductules: treatment with fulvestrant or with the aromatase inhibitor anastrozole. Whereas fulvestrant markedly increased Mmp7 and Spp1, and reduced Nptx1 mRNA levels, no changes were observed with anastrozole. Fulvestrant caused changes in epithelial morphology that were not seen with anastrozole. Fulvestrant shifted MMP7 immunolocalization in the epithelial cells from the supranuclear to the apical region; this effect was less pronounced with anastrozole. In vitro studies of (35)S-methionine incorporation showed that protein release was increased, whereas tissue protein content in the efferent ductules of fulvestrant-treated rats was decreased. Although fulvestrant markedly affected gene expression, no changes were observed on AP-1 and SP1 DNA-binding activity. The blockade of ESRs seems to be the major reason explaining the differences between both treatments. At least some of the effects of fulvestrant appear to result from compensatory mechanisms activated by the dramatic changes caused by ESR1 blockade.
Collapse
Affiliation(s)
- Gisele Renata Oliveira Gomes
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, Instituto Nacional de Farmacologia e Biologia Molecular, Vila Clementino, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
128
|
Huijbregts L, de Roux N. KISS1 is down-regulated by 17beta-estradiol in MDA-MB-231 cells through a nonclassical mechanism and loss of ribonucleic acid polymerase II binding at the proximal promoter. Endocrinology 2010; 151:3764-72. [PMID: 20534720 DOI: 10.1210/en.2010-0260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Kisspeptins are hypothalamic neuropeptides encoded by KISS1 and recently described as major regulators of GnRH release from hypothalamic neurons. Although 17beta-estradiol (E2)-induced up-regulation of KISS1 expression has been documented in anteroventral periventricular nucleus neurons, E2 down-regulates KISS1 expression in arcuate nucleus neurons via the estrogen receptor alpha by unknown molecular mechanisms. Because KISS1 was initially described as a metastasis inhibitor, notably in breast tumors, we used the MDA-MB-231 breast cancer cell line, which expresses high levels of KISS1, to characterize the molecular mechanism underlying KISS1 regulation by E2. E2 rapidly down-regulated endogenous KISS1 in a stable ERalpha-expressing MDA-MB-231 cell line. Promoter analysis revealed that E2 down-regulation was determined by a short 93-bp sequence devoid of estrogen response element and Sp1 sites. E2 down-regulation persisted with an ERalpha that was unable to bind DNA and in the presence of histone deacetylase inhibitor. In the absence of E2, unliganded ERalpha and RNA polymerase II (RNAPII) were present on the proximal promoter. E2 stimulation induced recruitment of ERalpha and loss of RNAPII at the proximal promoter. Along the gene body, total RNAPII amounts were similar in E2-treated and untreated cells, whereas the active form was significantly less abundant in E2-treated cells. Thus, E2-induced down-regulation of KISS1 is mediated by a pathway combining RNAPII loss at the proximal promoter and modulation of active RNAPII along the gene body, which is a novel mechanism in the complex process of E2-induced repression of gene expression.
Collapse
Affiliation(s)
- Lukas Huijbregts
- Avenir Team Genetic and Physiology of the Onset of Puberty, Institut National de la Santé et de laRecherche Médicale Unité 676, Hopital Robert Debré, and Université Paris Diderot, 75019 Paris, France
| | | |
Collapse
|
129
|
|
130
|
Gambino YP, Maymó JL, Pérez-Pérez A, Dueñas JL, Sánchez-Margalet V, Calvo JC, Varone CL. 17Beta-Estradiol Enhances Leptin Expression in Human Placental Cells Through Genomic and Nongenomic Actions1. Biol Reprod 2010; 83:42-51. [DOI: 10.1095/biolreprod.110.083535] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
131
|
Genome-wide analysis of estrogen receptor alpha DNA binding and tethering mechanisms identifies Runx1 as a novel tethering factor in receptor-mediated transcriptional activation. Mol Cell Biol 2010; 30:3943-55. [PMID: 20547749 DOI: 10.1128/mcb.00118-10] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nuclear receptor estrogen receptor alpha (ER alpha) controls the expression of hundreds of genes responsible for target cell phenotypic properties, but the relative importance of direct versus tethering mechanisms of DNA binding has not been established. In this first report, we examine the genome-wide chromatin localization of an altered-specificity mutant ER with a DNA binding domain deficient in binding to estrogen response element (ERE)-containing DNA (DBDmut ER) versus wild-type ER alpha. Using high-throughput sequencing of ER chromatin immunoprecipitations (ChIP-Seq) and mRNA transcriptional profiling, we show that direct ERE binding is required for most of (75%) estrogen-dependent gene regulation and 90% of hormone-dependent recruitment of ER to genomic binding sites. De novo motif analysis of the chromatin binding regions in MDA-MB-231 human breast cancer cells defined unique transcription factor profiles responsible for genes regulated through tethering versus direct ERE binding, with Runx motifs enriched in ER-tethered sites. We confirmed a role for Runx1 in mediating ER alpha genomic recruitment and regulation of tethering genes. Our findings delineate the contributions of direct receptor ERE binding versus binding through response elements for other transcription factors in chromatin localization and ER-dependent gene regulation, paradigms likely to underlie the gene regulatory actions of other nuclear receptors as well.
Collapse
|
132
|
Mandal S, Davie JR. Estrogen regulated expression of the p21 Waf1/Cip1 gene in estrogen receptor positive human breast cancer cells. J Cell Physiol 2010; 224:28-32. [PMID: 20301197 DOI: 10.1002/jcp.22078] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The cyclin-dependent kinase inhibitor protein p21(Waf1/Cip1) is a potent tumor suppressor. Here, we demonstrate that estradiol regulates the p21(Waf1/Cip1) gene. Estradiol induces p21(Waf1/Cip1) mRNA expression within 30-60 min independent of new protein synthesis in the estrogen receptor alpha (ER alpha) positive human breast cancer cell line MCF-7. Similar to other estradiol responsive promoters, the p21(Waf1/Cip1) upstream promoter region has several estrogen response element (ERE) half-sites nestled in AP-1 binding sites, which are positioned upstream to Sp1 binding sites. Using the chromatin immunoprecipitation (ChIP) assay, we show that estradiol stimulation resulted in the recruitment of transcription factors ER alpha, Sp1, and Sp3 to the p21(Waf1/Cip1) upstream promoter element. The Sp1 inhibitor mithramycin A abrogated Sp1, and to a lesser extent Sp3 binding, and markedly reduced the estradiol stimulated p21(Waf1/Cip1) gene expression. However, ER alpha binding was not affected in the mithramycin A and estradiol treated cells. On closer examination of the half-site ERE/AP-1 sites upstream to the Sp1 sites in a separate ChIP experiment, we found a pronounced association of ER alpha upon estradiol treatment compared to almost negligible binding of Sp1 or Sp3. Together these studies provide evidence that ER alpha is recruited to the half-site ERE/AP-1 sites in the p21(Waf1/Cip1) upstream promoter element. Although Sp1/Sp3 is not involved in the recruitment of ER alpha to the promoter, Sp1 is necessary for estrogen-induced p21(Waf1/Cip1) promoter activity.
Collapse
Affiliation(s)
- Soma Mandal
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
133
|
Li Y, Birnbaumer L, Teng CT. Regulation of ERRalpha gene expression by estrogen receptor agonists and antagonists in SKBR3 breast cancer cells: differential molecular mechanisms mediated by g protein-coupled receptor GPR30/GPER-1. Mol Endocrinol 2010; 24:969-80. [PMID: 20211987 PMCID: PMC2870941 DOI: 10.1210/me.2009-0148] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 02/01/2010] [Indexed: 12/21/2022] Open
Abstract
In selected tissues and cell lines, 17beta-estradiol (E2) regulates the expression of estrogen-related receptor alpha (ERRalpha), a member of the orphan nuclear receptor family. This effect is thought to be mediated by the estrogen receptor alpha (ERalpha). However in the ERalpha- and ERbeta-negative SKBR3 breast cancer cell line, physiological levels of E2 also stimulate ERRalpha expression. Here, we explored the molecular mechanism that mediates estrogen action in ER-negative breast cancer cells. We observed that E2, the ERalpha agonist, as well as the ERalpha antagonists ICI 182,780 and tamoxifen (TAM), a selective ER modulator, stimulate the transcriptional activity of the ERRalpha gene and increase the production of ERRalpha protein in SKBR3 cells. Moreover, the ERRalpha downstream target genes expression and cellular proliferation are also increased. We show further that the G protein-coupled receptor GPR30/GPER-1 (GPER-1) mediates these effects. The GPER-1 specific ligand G-1 mimics the actions of E2, ICI 182,780, and TAM on ERRalpha expression, and changing the levels of GPER-1 mRNA by overexpression or small interfering RNA knockdown affected the expression of ERRalpha accordingly. Utilizing inhibitors, we delineate a different downstream pathway for ER agonist and ER antagonist-triggered signaling through GPER-1. We also find differential histone acetylation and transcription factor recruitment at distinct nucleosomes of the ERRalpha promoter, depending on whether the cells are activated with E2 or with ER antagonists. These findings provide insight into the molecular mechanisms of GPER-1/ERRalpha-mediated signaling and may be relevant to what happens in breast cancer cells escaping inhibitory control by TAM.
Collapse
Affiliation(s)
- Yin Li
- Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | |
Collapse
|
134
|
Schultz DJ, Wickramasinghe NS, Ivanova MM, Isaacs SM, Dougherty SM, Imbert-Fernandez Y, Cunningham AR, Chen C, Klinge CM. Anacardic acid inhibits estrogen receptor alpha-DNA binding and reduces target gene transcription and breast cancer cell proliferation. Mol Cancer Ther 2010; 9:594-605. [PMID: 20197399 DOI: 10.1158/1535-7163.mct-09-0978] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anacardic acid (AnAc; 2-hydroxy-6-alkylbenzoic acid) is a dietary and medicinal phytochemical with established anticancer activity in cell and animal models. The mechanisms by which AnAc inhibits cancer cell proliferation remain undefined. AnAc 24:1(omega5) was purified from geranium (Pelargonium x hortorum) and shown to inhibit the proliferation of estrogen receptor alpha (ERalpha)-positive MCF-7 and endocrine-resistant LCC9 and LY2 breast cancer cells with greater efficacy than ERalpha-negative primary human breast epithelial cells, MCF-10A normal breast epithelial cells, and MDA-MB-231 basal-like breast cancer cells. AnAc 24:1(omega5) inhibited cell cycle progression and induced apoptosis in a cell-specific manner. AnAc 24:1(omega5) inhibited estradiol (E(2))-induced estrogen response element (ERE) reporter activity and transcription of the endogenous E(2) target genes pS2, cyclin D1, and cathepsin D in MCF-7 cells. AnAc 24:1(omega5) did not compete with E(2) for ERalpha or ERbeta binding, nor did AnAc 24:1(omega5) reduce ERalpha or ERbeta steady-state protein levels in MCF-7 cells; rather, AnAc 24:1(omega5) inhibited ER-ERE binding in vitro. Virtual screening with the molecular docking software Surflex evaluated AnAc 24:1(omega5) interaction with ERalpha ligand binding (LBD) and DNA binding (DBD) domains in conjunction with experimental validation. Molecular modeling revealed AnAc 24:1(omega5) interaction with the ERalpha DBD but not the LBD. Chromatin immunoprecipitation experiments revealed that AnAc 24:1(omega5) inhibited E(2)-ERalpha interaction with the endogenous pS2 gene promoter region containing an ERE. These data indicate that AnAc 24:1(omega5) inhibits cell proliferation, cell cycle progression, and apoptosis in an ER-dependent manner by reducing ER-DNA interaction and inhibiting ER-mediated transcriptional responses.
Collapse
Affiliation(s)
- David J Schultz
- Department of Biology, University of Louisville School of Medicine, Louisville, Kentucky 40292, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Zhang Z, Wang L, Mei M, Zhu Y, Du X, Lee C, Park I, Zhang J, Shi J. Both nongenomic and genomic effects are involved in estradiol's enhancing the phenotype of smooth muscle cells in cultured prostate stromal cells. Prostate 2010; 70:317-32. [PMID: 19862801 DOI: 10.1002/pros.21066] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Stromal smooth muscle cells (SMCs) play an important role in the pathogenesis and clinical symptom of benign prostatic hyperplasia. We had reported that estrogen enhances the phenotype of SMC in cultured prostate stromal cells (PRSCs). Here we further investigate the mechanism by which estrogen affects the differentiation of PRSCs. METHODS Primary cultured PRSCs were stimulated with E2 or BSA-E2. The mRNA level of SMC-specific genes, smoothelin, and SM-MHC were measured by qRT-PCR. The SM-MHC protein was measured by Western blot. The mRNA and protein levels of TGF-beta1 were measured by qRT-PCR and ELISA. The MAPK inhibitor PD98059, the estrogen receptor antagonist ICI182,780 and neutralizing antibody to TGF-beta1 were used to reveal the mechanism of estrogen effect. RESULTS E2 and BSA-E2 significantly up-regulate the expression of SMC-specific genes in PRSCs. Both forms of estrogen could increase the expression of TGF-beta1, which can be blocked by pre-treating with PD98059. Moreover, PD98059 and TGF-beta1 neutralizing antibody could abrogate the effect of BSA-E2 on cell differentiation. However, they could only inhibit part of E2-induced SMC phenotype enhancement. ICI182,780 could partially suppress the pro-differentiation effect of E2 but had no influence on the effect of BSA-E2. Combined treatment with ICI182,780 and PD98059 can completely abrogate the effect of E2. CONCLUSIONS Estrogen could promote the expression of TGF-beta1 in PRSCs through nongenomic activation of MAPK pathway, and in turn enhance the SMC phenotype. Besides for this nongenomic effect, estrogen can also enhance the SMC phenotype through classical genomic action.
Collapse
Affiliation(s)
- Zhisong Zhang
- Bioactive Materials Key Lab of Ministry of Education, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Mok SK, Chen WF, Lai WP, Leung PC, Wang XL, Yao XS, Wong MS. Icariin protects against bone loss induced by oestrogen deficiency and activates oestrogen receptor-dependent osteoblastic functions in UMR 106 cells. Br J Pharmacol 2010; 159:939-49. [PMID: 20128811 DOI: 10.1111/j.1476-5381.2009.00593.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Icariin may be the active ingredient in Herba Epimedii, a Chinese herb commonly used for treatment of osteoporosis. The present study aims to delineate the mechanism(s) by which icariin prevents bone loss after ovariectomy (OVX) in vivo and stimulates osteoblastic functions in vitro. EXPERIMENTAL APPROACH Ovariectomized or sham-operated C57BL/6 mice were treated with vehicle, 17beta-oestradiol or icariin for 6 weeks. Total and trabecular bome mineral density (BMD) as well as polar stress-strain index of distal femur were measured by peripheral computed tomography. The mRNA expressions of OPG and RANKL in tibia were studied by RT-PCR. Interactions between the oestrogen receptor (ER) antagonist ICI182,780 and icariin were studied in UMR 106 cells. The functional transactivation of ERalpha and ERbeta as well as ERalpha phosphorylation by icariin were also assessed. KEY RESULTS Icariin suppressed the loss of bone mass and strength in distal femur and increased the mRNA expression ratio of OPG/RANKL in tibia, following OVX. Icariin increased ER-dependent cell proliferation, alkaline phosphatase (ALP) activity, gene expression of OPG and the OPG/RANKL ratio in UMR 106 cells. Icariin did not activate ERE-luciferase activity in UMR 106 cells, via the ERalpha or the ERbeta-mediated pathway, but it did increase ERalpha phosphorylation at Ser118. CONCLUSIONS AND IMPLICATIONS Our results indicate that icariin exerts anabolic effects in bone possibly by activating ER in a ligand-independent manner. Its ability to prevent OVX-induced bone loss without inducing uterotrophic effects supports its use as an alternative regimen for management of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Sao-Keng Mok
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
137
|
Sisci D, Middea E, Morelli C, Lanzino M, Aquila S, Rizza P, Catalano S, Casaburi I, Maggiolini M, Andò S. 17β-estradiol enhances α(5) integrin subunit gene expression through ERα-Sp1 interaction and reduces cell motility and invasion of ERα-positive breast cancer cells. Breast Cancer Res Treat 2010; 124:63-77. [PMID: 20052536 DOI: 10.1007/s10549-009-0713-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 12/23/2009] [Indexed: 11/26/2022]
Abstract
In breast tumors the expression of estrogen receptor alpha (ERα) is known to be associated with a more favorable prognosis. ERα expression has been reported to reduce the metastatic potential of breast cancer cells. Recently, we have observed that extracellular matrix proteins activate ERα and that both liganded and unliganded receptor modulate cell invasiveness acting at nuclear level. To explain the mechanisms by which ERα regulates cell adhesion, we have evaluated the expression of α(5)β(1) integrin, prevalently expressed in stationary cells, in response to 17β-estradiol (E2). Here we show that E2/ERα increases the expression of integrin α(5)β(1) through Sp1-mediated binding to a GC-rich region located upstream of an ERE half-site in the 5' flanking region of the α(5) gene forming a ternary ERα-Sp1-DNA complex. Estrogen responsiveness of the α(5) gene promoter, as observed in HeLa cells, underlies a general mechanism of regulation which is not strictly linked to the cell type. Our data reveal novel insight into the molecular mechanisms sustaining the reduced invasiveness of ERα expressing cells demonstrating that α(5)β(1) integrin expression is related to the maintenance of the stationary status of the cells, counteracting E2/ERα capability to enhance breast cancer cell migration and invasion.
Collapse
Affiliation(s)
- Diego Sisci
- Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Mueck AO, Seeger H, Shapiro S. Risk of breast cancer during hormone replacement therapy: mechanisms. Horm Mol Biol Clin Investig 2010; 3:329-39. [DOI: 10.1515/hmbci.2010.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 07/26/2010] [Indexed: 11/15/2022]
Abstract
AbstractRegarding estrogen replacement therapy, two main mechanisms have to be considered for it to be discussed as a potential carcinogen in the breast, and also considering the World Health Organization definition of estrogens and estrogen/progestogen combinations as “carcinogenic”: (i) the proliferative/apoptotic effects on already pre-existing estrogen-sensitive cancer cells and (ii) the production of possible genotoxic estrogen metabolites. By addition of the progestogen component, as is usual in non-hysterectomized women, both mechanisms can lead to an increased risk compared to estrogenonly therapy. The detailed mechanisms underlying the development of the benign breast epithelial cell into clinically relevant breast cancer cells are very complicated. Based on these mechanisms, the following simplified summary of the main steps explains that: (i) an increased risk cannot be excluded, (ii) especially when estrogens are combined with progestogens, but (iii) there are differences between the preparations used in therapy; (iv) the risk seems to be very rare, needing very special cellular and extracellular conditions, (v) and could even be decreased in special situations of estrogen therapy. It is concluded that when critically reviewed, an increased risk of breast cancer during hormone replacement therapy cannot be excluded in very rare cases. Definitive mechanistic evidence for a possible causal relationship with carcinogenesis still remains open.
Collapse
|
139
|
Nott SL, Huang Y, Kalkanoglu A, Harper K, Chen M, Paoni SF, Fenton BM, Muyan M. Designer monotransregulators provide a basis for a transcriptional therapy for de novo endocrine-resistant breast cancer. Mol Med 2009; 16:10-8. [PMID: 19946606 DOI: 10.2119/molmed.2009.00107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 11/15/2009] [Indexed: 02/05/2023] Open
Abstract
The main circulating estrogen hormone 17beta-estradiol (E2) contributes to the initiation and progression of breast cancer. Estrogen receptors (ERs), as transcription factors, mediate the effects of E2. Ablation of the circulating E2 and/or prevention of ER functions constitute approaches for ER-positive breast cancer treatments. These modalities are, however, ineffective in de novo endocrine-resistant breast neoplasms that do not express ERs. The interaction of E2-ERs with specific DNA sequences, estrogen responsive elements (EREs), of genes constitutes one genomic pathway necessary for cellular alterations. We herein tested the prediction that specific regulation of ERE-driven genes by an engineered monomeric and constitutively active transcription factor, monotransregulator, provides a basis for the treatment of ER-negative breast cancer. Using adenovirus infected ER-negative MDA-MB-231 cells derived from a breast adenocarcinoma, we found that the monotransregulator, but not the ERE-binding defective counterpart, repressed cellular proliferation and motility, and induced apoptosis through expression of genes that required ERE interactions. Similarly, the monotransregulator suppressed the growth of ER-negative BT-549 cells derived from a breast-ductal carcinoma. Moreover, the ERE-binding monotransregulator repressed xenograft tumor growth in a nude mice model. Thus, specific regulation of genes bearing EREs could offer a therapeutic approach for de novo endocrine-resistant breast cancers.
Collapse
Affiliation(s)
- Stephanie L Nott
- Department of Biochemistry and Biophysics, University of Rochester Medical School, Rochester, New York, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
Activation of estrogen receptor alpha (ERalpha) results in both induction and repression of gene transcription; while mechanistic details of estrogen induction are well described, details of repression remain largely unknown. We characterized several ERalpha-repressed targets and examined in detail the mechanism for estrogen repression of Reprimo (RPRM), a cell cycle inhibitor. Estrogen repression of RPRM is rapid and robust and requires a tripartite interaction between ERalpha, histone deacetylase 7 (HDAC7), and FoxA1. HDAC7 is the critical HDAC needed for repression of RPRM; it can bind to ERalpha and represses ERalpha's transcriptional activity--this repression does not require HDAC7's deacetylase activity. We further show that the chromatin pioneer factor FoxA1, well known for its role in estrogen induction of genes, is recruited to the RPRM promoter, is necessary for repression of RPRM, and interacts with HDAC7. Like other FoxA1 recruitment sites, the RPRM promoter is characterized by H3K4me1/me2. Estrogen treatment causes decreases in H3K4me1/me2 and release of RNA polymerase II (Pol II) from the RPRM proximal promoter. Overall, these data implicate a novel role for HDAC7 and FoxA1 in estrogen repression of RPRM, a mechanism which could potentially be generalized to many more estrogen-repressed genes and hence be important in both normal physiology and pathological processes.
Collapse
|
141
|
Mahmoodzadeh S, Dworatzek E, Fritschka S, Pham TH, Regitz-Zagrosek V. 17beta-Estradiol inhibits matrix metalloproteinase-2 transcription via MAP kinase in fibroblasts. Cardiovasc Res 2009; 85:719-28. [PMID: 19861308 PMCID: PMC2819834 DOI: 10.1093/cvr/cvp350] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AIMS Female sex and sex hormones contribute to cardiac remodelling. 17beta-estradiol (E2) is involved in the modulation of extracellular matrix composition and function. Here, we analysed the effect of E2 on matrix metalloproteinase (MMP)-2 gene expression and studied the underlying molecular mechanisms in rat cardiac fibroblasts and in a human fibroblast cell line. METHODS AND RESULTS In adult rat cardiac fibroblasts, E2 significantly decreased MMP-2 gene expression in an estrogen receptor (ER)-dependent manner. Transient transfection experiments of human MMP-2 (hMMP-2) promoter deletion constructs in a human fibroblast cell line revealed a regulatory region between -324 and -260 bp that is involved in E2/ERalpha-mediated repression of hMMP-2 gene transcription. Electrophoretic mobility shift assays (EMSA) and supershift analysis demonstrated the binding of transcription factor Elk-1 within this promoter region. Elk-1 was phosphorylated by E2 via the mitogen-activated protein kinase (MAPK) signalling pathway as shown by western blotting. Treatment of cells with the MAPK inhibitor PD98059 blocked the E2-dependent repression of hMMP-2 promoter activity as well as the endogenous MMP-2 mRNA levels in both human fibroblast cells and rat cardiac fibroblasts. CONCLUSION E2 inhibits MMP-2 expression via the ER and the MAPK pathway in rat cardiac fibroblasts and in a human fibroblast cell line. These mechanisms may contribute to sex-specific differences in fibrotic processes that are observed in human heart and other diseases.
Collapse
Affiliation(s)
- Shokoufeh Mahmoodzadeh
- Institute of Gender in Medicine , Charite-Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany
| | | | | | | | | |
Collapse
|
142
|
Sakamoto T, Horiguchi H, Oguma E, Kayama F. Effects of diverse dietary phytoestrogens on cell growth, cell cycle and apoptosis in estrogen-receptor-positive breast cancer cells. J Nutr Biochem 2009; 21:856-64. [PMID: 19800779 DOI: 10.1016/j.jnutbio.2009.06.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 04/13/2009] [Accepted: 06/23/2009] [Indexed: 01/08/2023]
Abstract
Phytoestrogens have attracted attention as being safer alternatives to hormone replacement therapy (HRT) and as chemopreventive reagents for breast cancer because dietary soy isoflavone intake has been correlated with reduction in risk. To identify safe and effective phytoestrogen candidates for HRT and breast cancer prevention, we investigated the effects of daidzein, genistein, coumestrol, resveratrol and glycitein on cell growth, cell cycle, cyclin D1 expression, apoptosis, Bcl-2/Bax expression ratio and p53-dependent or NF-kappaB-dependent transcriptional activity in MCF-7 breast cancer cells. Phytoestrogens, except for glycitein, significantly enhanced estrogen-response-element-dependent transcriptional activity up to a level similar to that of 17beta-estradiol (E(2)). E(2) increased cell growth significantly, coumestrol increased cell growth moderately, and resveratrol and glycitein reduced cell growth. Phytoestrogens, except for glycitein, stimulated the promotion of cells to G(1)/S transition in cell cycle analysis, similar to E(2). This stimulation was accompanied by transient up-regulation of cyclin D1. While genistein, resveratrol and glycitein all increased apoptosis and reduced the Bcl-2/Bax ratio, resveratrol reduced this ratio more than either genistein or glycitein. Moreover, resveratrol significantly enhanced p53-dependent transcriptional activity, but slightly reduced NF-kappaB-dependent transcriptional activity. On knockdown analysis, genistein, resveratrol and glycitein all reduced the Bcl-2/Bax ratio in the presence of apoptosis-inducing stimuli, and estrogen receptor (ER) alpha silencing had no effect on these reductions. In contrast, in the absence of apoptosis-inducing stimuli, only resveratrol reduced the ratio, and ERalpha silencing abolished this reduction. Thus, resveratrol might be the most promising candidate for HRT and chemoprevention of breast cancer due to its estrogenic activity and high antitumor activity.
Collapse
Affiliation(s)
- Takako Sakamoto
- Division of Environmental Medicine, Center for Community Medicine, Jichi Medical University, Shimotsuke City, Tochigi, Japan.
| | | | | | | |
Collapse
|
143
|
Regulation of Kiss1 and dynorphin gene expression in the murine brain by classical and nonclassical estrogen receptor pathways. J Neurosci 2009; 29:9390-5. [PMID: 19625529 DOI: 10.1523/jneurosci.0763-09.2009] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Kisspeptin is a product of the Kiss1 gene and is expressed in the forebrain. Neurons that express Kiss1 play a crucial role in the regulation of pituitary luteinizing hormone secretion and reproduction. These neurons are the direct targets for the action of estradiol-17beta (E(2)), which acts via the estrogen receptor alpha isoform (ER alpha) to regulate Kiss1 expression. In the arcuate nucleus (Arc), where the dynorphin gene (Dyn) is expressed in Kiss1 neurons, E(2) inhibits the expression of Kiss1 mRNA. However, E(2) induces the expression of Kiss1 in the anteroventral periventricular nucleus (AVPV). The mechanism for differential regulation of Kiss1 in the Arc and AVPV by E(2) is unknown. ER alpha signals through multiple pathways, which can be categorized as either classical, involving the estrogen response element (ERE), or nonclassical, involving ERE-independent mechanisms. To elucidate the molecular basis for the action of E(2) on Kiss1 and Dyn expression, we studied the effects of E(2) on Kiss1 and Dyn mRNAs in the brains of mice bearing targeted alterations in the ER alpha signaling pathways. We found that stimulation of Kiss1 expression by E(2) in the AVPV and inhibition of Dyn in the Arc required an ERE-dependent pathway, whereas the inhibition of Kiss1 expression by E(2) in the Arc involved ERE-independent mechanisms. Thus, distinct ER alpha signaling pathways can differentially regulate the expression of identical genes across different brain regions, and E(2) can act within the same neuron through divergent ER alpha signaling pathways to regulate different neurotransmitter genes.
Collapse
|
144
|
Chen JQ, Cammarata PR, Baines CP, Yager JD. Regulation of mitochondrial respiratory chain biogenesis by estrogens/estrogen receptors and physiological, pathological and pharmacological implications. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1540-70. [PMID: 19559056 DOI: 10.1016/j.bbamcr.2009.06.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 12/21/2022]
Abstract
There has been increasing evidence pointing to the mitochondrial respiratory chain (MRC) as a novel and important target for the actions of 17beta-estradiol (E(2)) and estrogen receptors (ER) in a number of cell types and tissues that have high demands for mitochondrial energy metabolism. This novel E(2)-mediated mitochondrial pathway involves the cooperation of both nuclear and mitochondrial ERalpha and ERbeta and their co-activators on the coordinate regulation of both nuclear DNA- and mitochondrial DNA-encoded genes for MRC proteins. In this paper, we have: 1) comprehensively reviewed studies that reveal a novel role of estrogens and ERs in the regulation of MRC biogenesis; 2) discussed their physiological, pathological and pharmacological implications in the control of cell proliferation and apoptosis in relation to estrogen-mediated carcinogenesis, anti-cancer drug resistance in human breast cancer cells, neuroprotection for Alzheimer's disease and Parkinson's disease in brain, cardiovascular protection in human heart and their beneficial effects in lens physiology related to cataract in the eye; and 3) pointed out new research directions to address the key questions in this important and newly emerging area. We also suggest a novel conceptual approach that will contribute to innovative regimens for the prevention or treatment of a wide variety of medical complications based on E(2)/ER-mediated MRC biogenesis pathway.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|
145
|
Nichols M, Cheng P, Liu Y, Kanterewicz B, Hershberger PA, McCarty KS. Breast cancer-derived M543V mutation in helix 12 of estrogen receptor alpha inverts response to estrogen and SERMs. Breast Cancer Res Treat 2009; 120:761-8. [PMID: 19526339 DOI: 10.1007/s10549-009-0437-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Accepted: 06/03/2009] [Indexed: 02/06/2023]
Abstract
We have isolated from human breast cancers several mutations in the Helix 12 component of activation function 2 (AF-2) in the estrogen receptor alpha (ERalpha). We used a novel approach to detect changes in the hormone-binding domain of ERalpha, based on the evidence that antiestrogens, such as 4-hydroxytamoxifen (ZOHT) and ICI 182,780, block the function of ERalpha by binding and folding the AF-2 transcriptional domain in a way that inhibits its association with coactivator proteins. We have identified a Helix 12 mutation, M543V, which leads to greater ERalpha transcription with ZOHT and other antiestrogens (including 1,1-dichloro-2,2,3-triarylcyclopropanes, DTACs) than with 17-beta estradiol (E2). We also found an independent mutation at the same position, M543I, which did not show this inverted ligand phenotype. In comparison to further Helix 12 mutations made in vitro, it appears that relative hydrophobicity of the amino acid side chains on the inner face of Helix 12 is key to maintaining the transcriptionally active, agonist conformation with bound E2. This active conformation can be induced, resulting in increased transcription, by adding excess p160 coactivator AIB1 in transcriptional assays with E2-bound receptors, while the ZOHT-bound receptors were not further activated by AIB1. Other experiments show that the cross talk between ERalpha and AP-1 protein from AP-1-binding sites is not dependent on Helix 12 integrity. We show that two alleles containing a proline substitution in Helix 12 that inactivate AF-2 function of ERalpha at EREs have little negative effect on function through AP-1 elements, supporting a prominent role for the N-terminal AF-1 of ERalpha in AP-1/ERalpha transcriptional cross talk.
Collapse
Affiliation(s)
- Mark Nichols
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | |
Collapse
|
146
|
Abstract
This review emphasizes the relationship of breast cancer, estrogen receptor and ligands, especially the centrality of the estrogen receptor, which mediates on one hand the hormone-induced gene transcription and on the other hand the anti-estrogen action against breast cancer. The characterization of the estrogen receptor ligand-binding domain co-crystallized with agonists or antagonists provided a molecular basis to gain an insight into the regulation of estrogen receptor and, thereby, to describe the mechanism of the hormone therapy in treating breast cancer.
Collapse
Affiliation(s)
- Zhenlin Bai
- Institute of Pharmacy, Freie Universität Berlin, Knigin-Luise-Strasse 2 + 4, Berlin, Germany
| | | |
Collapse
|
147
|
Chen JQ, Brown TR, Russo J. Regulation of energy metabolism pathways by estrogens and estrogenic chemicals and potential implications in obesity associated with increased exposure to endocrine disruptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1128-43. [PMID: 19348861 DOI: 10.1016/j.bbamcr.2009.03.009] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 03/26/2009] [Accepted: 03/27/2009] [Indexed: 10/20/2022]
Abstract
The prevalence of obesity among children, adolescents and adults has been dramatically increasing worldwide during the last several decades. The obesity epidemic has been recognized as one of the major global health problems, because its health hazard is linked to a number of common diseases including breast and prostate cancers. Obesity is caused by combination of genetic and environmental factors. While genetic contribution to obesity has been known to be significant, the genetic factors remain relatively unchanged. Recent studies have highlighted the involvement of environmental "obesogens", i.e. the xenobiotic chemicals that can disrupt the normal development and homeostatic control over adipogenesis and energy balance. Several lines of evidence suggest that increasing exposure to chemicals with endocrine-disrupting activities (endocrine-disrupting chemicals, EDCs) contributes to the increased obesity. The cellular and molecular mechanisms underlying obesogen-associated obesity are just now being appreciated. In this paper, we comprehensively reviewed current knowledge about the role of estrogen receptors alpha and beta (ERalpha and ERbeta) in regulation of energy metabolism pathways, including glucose transport, glycolysis, tricarboxylic acid (TCA) cycle, mitochondrial respiratory chain (MRC), adenosine nucleotide translocator (ANT) and fatty acid beta-oxidation and synthesis, by estrogens; and then examined the disturbance of E(2)/ER-mediated energy metabolism pathways by environmental obesogens; and finally, we discussed the potential implications of disturbance of energy metabolism pathways by obesogens in obesity and pointed out several key aspects of this area that need to be further explored. A better understanding of the cellular and molecular mechanisms underlying obesogen-associated obesity will lead to new approaches for slow down and/or prevention of the increased trend of obesity associated with exposure to obesogens.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| | | | | |
Collapse
|
148
|
Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol 2009; 47:787-95. [DOI: 10.1016/j.fct.2009.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 12/12/2008] [Accepted: 01/05/2009] [Indexed: 11/20/2022]
|
149
|
Nott SL, Huang Y, Li X, Fluharty BR, Qiu X, Welshons WV, Yeh S, Muyan M. Genomic responses from the estrogen-responsive element-dependent signaling pathway mediated by estrogen receptor alpha are required to elicit cellular alterations. J Biol Chem 2009; 284:15277-88. [PMID: 19321454 DOI: 10.1074/jbc.m900365200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Estrogen (E2) signaling is conveyed by the transcription factors estrogen receptor (ER) alpha and beta. ERs modulate the expression of genes involved in cellular proliferation, motility, and death. The regulation of transcription by E2-ERalpha through binding to estrogen-responsive elements (EREs) in DNA constitutes the ERE-dependent signaling pathway. E2-ERalpha also modulates gene expression by interacting with transregulators bound to cognate DNA-regulatory elements, and this regulation is referred to as the ERE-independent signaling pathway. The relative importance of the ERE-independent pathway in E2-ERalpha signaling is unclear. To address this issue, we engineered an ERE-binding defective ERalpha mutant (ERalpha(EBD)) by changing residues in an alpha-helix of the protein involved in DNA binding to render the receptor functional only through the ERE-independent signaling pathway. Using recombinant adenovirus-infected ER-negative MDA-MB-231 cells derived from a breast adenocarcinoma, we found that E2-ERalpha(EBD) modulated the expression of a subset of ERalpha-responsive genes identified by microarrays and verified by quantitative PCR. However, E2-ERalpha(EBD) did not affect cell cycle progression, cellular growth, death, or motility in contrast to E2-ERalpha.ERalpha(EBD) in the presence of E2 was also ineffective in inducing phenotypic alterations in ER-negative U-2OS cells derived from an osteosarcoma. E2-ERalpha, on the other hand, effectively repressed growth in this cell line. Our findings suggest that genomic responses from the ERE-dependent signaling pathway are required for E2-ERalpha to induce alterations in cellular responses.
Collapse
Affiliation(s)
- Stephanie L Nott
- Department of Biochemistry and Biophysics, University of Rochester Medical School, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Kumar P, Kamat A, Mendelson CR. Estrogen receptor alpha (ERalpha) mediates stimulatory effects of estrogen on aromatase (CYP19) gene expression in human placenta. Mol Endocrinol 2009; 23:784-93. [PMID: 19299445 DOI: 10.1210/me.2008-0371] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A 246-bp region upstream of placenta-specific exon I.1 of the human aromatase (hCYP19) gene mediates placenta-specific, developmental, and O(2) regulation of expression. In this study, trophoblast differentiation and associated induction of CYP19 expression were prevented when cytotrophoblasts were cultured in phenol red-free medium containing charcoal-stripped serum or with the estrogen receptor (ER) antagonist, ICI 182,780, suggesting a stimulatory role of estrogen/ER. ERalpha protein was expressed in human trophoblasts and increased during syncytiotrophoblast differentiation, whereas ERbeta was undetectable. Mutational analysis revealed that an estrogen response element-like sequence (ERE-LS) at -208 bp is required for inductive effects of estradiol/ERalpha on hCYP19I.1 promoter activity in transfected COS-7 cells. Increased binding of syncytiotrophoblast compared with cytotrophoblast nuclear proteins to the ERE-LS was observed in vitro; however, ERalpha antibodies failed to supershift the complex and in vitro-transcribed/translated ERalpha did not bind. Nonetheless, chromatin immunoprecipitation assays in cultured trophoblasts revealed recruitment of endogenous ERalpha to the -255- to -155-bp region containing the ERE-LS before induction of hCYP19 expression; this was inhibited by ICI 182,780. Chromatin immunoprecipitation also revealed increased acetylated histone H3(K9/14) and decreased methylated histone H3(K9) associated with this region during trophoblast differentiation. These modifications were prevented when trophoblasts were incubated with ICI 182,780, suggesting that ERalpha recruitment to the -255- to -155-bp region promotes histone modifications leading to increased hCYP19 transcription. Thus, during trophoblast differentiation, estrogen/ERalpha exerts a positive feedback role, which promotes permissive histone modifications that are associated with induction of hCYP19 gene transcription.
Collapse
Affiliation(s)
- Premlata Kumar
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, 75390-9038, USA
| | | | | |
Collapse
|