1
|
Amenyogbe E, Chen G, Wang Z, Lu X, Lin M, Lin AY. A Review on Sex Steroid Hormone Estrogen Receptors in Mammals and Fish. Int J Endocrinol 2020; 2020:5386193. [PMID: 32089683 PMCID: PMC7029290 DOI: 10.1155/2020/5386193] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/04/2019] [Accepted: 11/19/2019] [Indexed: 12/26/2022] Open
Abstract
Steroid hormones play essential roles in the reproductive biology of vertebrates. Estrogen exercises its effect through estrogen receptors and is not only a female reproductive hormone but acts virtually in all vertebrates, including fish, and is involved in the physiological and pathological states in all males and females. Estrogen has been implicated in mandible conservation and circulatory and central nervous systems as well as the reproductive system. This review intended to understand the structure, function, binding affinities, and activations of estrogens and estrogen receptors and to discuss the understanding of the role of sex steroid hormone estrogen receptors in mammals and fish.
Collapse
Affiliation(s)
- Eric Amenyogbe
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| | - Gang Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| | - Zhongliang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| | - Xiaoying Lu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| | - Mingde Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| | - Ai Ying Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524025, China
- Guangdong Provincial Key Laboratory of Aquaculture in the South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Laboratory of Fish Aquaculture, Zhanjiang 524025, China
| |
Collapse
|
2
|
Gaudet HM, Christensen E, Conn B, Morrow S, Cressey L, Benoit J. Methylmercury promotes breast cancer cell proliferation. Toxicol Rep 2018; 5:579-584. [PMID: 29868453 PMCID: PMC5984200 DOI: 10.1016/j.toxrep.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 05/02/2018] [Accepted: 05/13/2018] [Indexed: 10/26/2022] Open
Abstract
CONTEXT Metalloestrogens are small ionic metals that activate the estrogen receptor (ER). Studies have shown that when metalloestrogens bind to the ER, there is an increase in transcription and expression of estrogen-regulated genes, which induces proliferation of estrogen-dependent breast cancer. Methylmercury (MeHg), a metalloestrogen, is present in the environment and is toxic at moderate to high concentrations. However, at lower concentrations MeHg may promote the proliferation of ER-positive breast cancers and protect cells against pro-apoptotic signals. OBJECTIVE To investigate the effects of MeHg treatment on breast cancer cells in vitro. MATERIALS AND METHODS MCF7 breast cancer cells were treated with concentrations of MeHg ranging from 1 nM to 100 mM. Hg analysis was used to quantify intracellular mercury concentrations. Cell proliferation and apoptosis were determined by cell counting and Annexin-V staining, respectively. RESULTS We defined a protocol that maximizes cellular exposure to mercury. Treatment of human ER-positive breast cancer cells with 1 nM MeHg promoted proliferation, while treatment with a concentration of 100 nM induced apoptosis. DISCUSSION AND CONCLUSIONS Clarifying the effects of MeHg on breast cancer will improve our understanding of how environmental toxins affect tumor progression and may lead to the development of future therapeutic strategies.
Collapse
|
3
|
Reuquen P, Guajardo-Correa E, Oróstica ML, Curotto C, Parada-Bustamante A, Cardenas H, Orihuela PA. Prolactin gene expression in the pituitary of rats subjected to vaginocervical stimulation requires Erk-1/2 signaling. Reprod Biol 2017; 17:357-362. [DOI: 10.1016/j.repbio.2017.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 09/21/2017] [Accepted: 10/03/2017] [Indexed: 01/28/2023]
|
4
|
Koong LY, Watson CS. Rapid, nongenomic signaling effects of several xenoestrogens involved in early- vs. late-stage prostate cancer cell proliferation. ACTA ACUST UNITED AC 2015. [DOI: 10.4161/23273747.2014.995003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Luke Y Koong
- Biochemistry & Molecular Biology Department; University of Texas Medical Branch; Galveston, TX USA
| | | |
Collapse
|
5
|
Viñas R, Goldblum RM, Watson CS. Rapid estrogenic signaling activities of the modified (chlorinated, sulfonated, and glucuronidated) endocrine disruptor bisphenol A. ACTA ACUST UNITED AC 2014. [DOI: 10.4161/endo.25411] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
6
|
Viñas R, Jeng YJ, Watson CS. Non-genomic effects of xenoestrogen mixtures. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2012; 9:2694-714. [PMID: 23066391 PMCID: PMC3447581 DOI: 10.3390/ijerph9082694] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/09/2012] [Accepted: 07/17/2012] [Indexed: 12/13/2022]
Abstract
Xenoestrogens (XEs) are chemicals derived from a variety of natural and anthropogenic sources that can interfere with endogenous estrogens by either mimicking or blocking their responses via non-genomic and/or genomic signaling mechanisms. Disruption of estrogens' actions through the less-studied non-genomic pathway can alter such functional end points as cell proliferation, peptide hormone release, catecholamine transport, and apoptosis, among others. Studies of potentially adverse effects due to mixtures and to low doses of endocrine-disrupting chemicals have recently become more feasible, though few so far have included actions via the non-genomic pathway. Physiologic estrogens and XEs evoke non-monotonic dose responses, with different compounds having different patterns of actions dependent on concentration and time, making mixture assessments all the more challenging. In order to understand the spectrum of toxicities and their mechanisms, future work should focus on carefully studying individual and mixture components across a range of concentrations and cellular pathways in a variety of tissue types.
Collapse
Affiliation(s)
- René Viñas
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | | | |
Collapse
|
7
|
Jenkins S, Betancourt AM, Wang J, Lamartiniere CA. Endocrine-active chemicals in mammary cancer causation and prevention. J Steroid Biochem Mol Biol 2012; 129:191-200. [PMID: 21729753 DOI: 10.1016/j.jsbmb.2011.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/31/2011] [Accepted: 06/01/2011] [Indexed: 01/11/2023]
Abstract
Endocrine-active chemicals alter or mimic physiological hormones. These compounds are reported to originate from a wide variety of sources, and recent studies have shown widespread human exposure to several of these compounds. Given the role of the sex steroid hormone, estradiol, in human breast cancer causation, endocrine-active chemicals which interfere with estrogen signaling constitute one potential factor contributing to the high incidence of breast cancer. Thus, the aim of this review is to examine several common endocrine-active chemicals and their respective roles in breast cancer causation or prevention. The plastic component, bisphenol A (BPA), the synthetic estrogen, diethylstilbestrol (DES), the by-product of organic combustion, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the soy component, genistein, and the red grape phytoalexin, resveratrol, have some degree of structural similarities to each other and estradiol. However, despite these structural similarities, the in vitro and in vivo properties of each of these chemicals vary greatly in terms of breast cancer causation and prevention. Early life exposure to BPA and DES increases rodent susceptibility to chemically induced mammary carcinogenesis, presumably through retardation of normal mammary gland maturation and/or disrupting the ratio of cell proliferation and apoptosis in the mammary gland. On the other hand, early exposures to genistein and resveratrol protect rodents against chemically induced and spontaneous mammary cancers. This is reported to occur through the ability of genistein and resveratrol to accelerate mammary gland maturation. Interestingly, TCDD, which is the most structurally dissimilar to the above chemicals and functions as an anti-estrogen, also increases chemically induced mammary carcinogenesis through retardation of mammary gland maturation. This article is part of a Special Issue entitled 'Endocrine disruptors'.
Collapse
Affiliation(s)
- Sarah Jenkins
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
8
|
Watson CS, Jeng YJ, Guptarak J. Endocrine disruption via estrogen receptors that participate in nongenomic signaling pathways. J Steroid Biochem Mol Biol 2011; 127:44-50. [PMID: 21300151 PMCID: PMC3106143 DOI: 10.1016/j.jsbmb.2011.01.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 01/27/2011] [Accepted: 01/30/2011] [Indexed: 12/21/2022]
Abstract
When inappropriate (non-physiologic) estrogens affect organisms at critical times of estrogen sensitivity, disruption of normal endocrine functions can result. Non-physiologic estrogen mimetics (environmental, dietary, and pharmaceutical) can signal rapidly and potently via the membrane versions of estrogen receptors, as can physiologic estrogens. Both physiologic and non-physiologic estrogens activate multiple signaling pathways, leading to altered cellular functions (e.g. peptide release, cell proliferation or death, transport). Xenoestrogens' mimicry of physiologic estrogens is imperfect. When superimposed, xenoestrogens can alter endogenous estrogens' signaling and thereby disrupt normal signaling pathways, leading to malfunctions in many tissue types. Though these xenoestrogen actions occur rapidly via nongenomic signaling pathways, they can be sustained with continuing ligand stimulation, combinations of ligands, and signaling that perpetuates downstream, eventually also impinging on genomic regulation by controlling the activation state of transcription factors. Because via these pathways estrogens and xenoestrogens cause nonmonotonic stimulation patterns, they must be carefully tested for activity and toxicity over wide dose ranges. Nongenomic actions of xenoestrogens in combination with each other, and with physiologic estrogens, are still largely unexplored from these mechanistic perspectives.
Collapse
Affiliation(s)
- Cheryl S. Watson
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555-0645, USA
| | - Yow-Juin Jeng
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555-0645, USA
| | - Jutatip Guptarak
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555-0645, USA
| |
Collapse
|
9
|
Tong JS, Zhang QH, Wang ZB, Li S, Yang CR, Fu XQ, Hou Y, Wang ZY, Sheng J, Sun QY. ER-α36, a novel variant of ER-α, mediates estrogen-stimulated proliferation of endometrial carcinoma cells via the PKCδ/ERK pathway. PLoS One 2010; 5:e15408. [PMID: 21079811 PMCID: PMC2973969 DOI: 10.1371/journal.pone.0015408] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/09/2010] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Recently, a variant of ER-α, ER-α36 was identified and cloned. ER-α36 lacks intrinsic transcription activity and mainly mediates non-genomic estrogen signaling. The purpose of this study was to investigate the function and the underlying mechanisms of ER-α36 in growth regulation of endometrial Ishikawa cancer cells. METHODS The cellular localization of ER-α36 and ER-α66 were determined by immunofluorescence in the Ishikawa cells. Ishikawa endometrial cancer control cells transfected with an empty expression vector, Ishikawa cells with shRNA knockdown of ER-α36 (Ishikawa/RNAiER36) and Ishikawa cells with shRNA knockdown of ER-α66 (Ishikawa/RNAiER66) were treated with E2 and E2-conjugated to bovine serum albumin (E2-BSA, membrane impermeable) in the absence and presence of different kinase inhibitors HBDDE, bisindolylmaleimide, rottlerin, H89 and U0126. The phosphorylation levels of signaling molecules and cyclin D1/cdk4 expression were examined with Western blot analysis and cell growth was monitored with the MTT assay. RESULTS Immunofluorescence staining of Ishikawa cells demonstrated that ER-α36 was expressed mainly on the plasma membrane and in the cytoplasm, while ER-α66 was predominantly localized in the cell nucleus. Both E2 and E2-BSA rapidly activated PKCδ not PKCα in Ishikawa cells, which could be abrogated by ER-α36 shRNA expression. E2-and E2-BSA-induced ERK phosphorylation required ER-α36 and PKCδ. However, only E2 was able to induce Camp-dependent protein kinase A (PKA) phosphorylation. Furthermore, E2 enhances cyclin D1/cdk4 expression via ER-α36. CONCLUSION E2 activates the PKCδ/ERK pathway and enhances cyclin D1/cdk4 expression via the membrane-initiated signaling pathways mediated by ER-α36, suggesting a possible involvement of ER-α36 in E2-dependent growth-promoting effects in endometrial cancer cells.
Collapse
Affiliation(s)
- Jing-Shan Tong
- College of Life Sciences, Jilin University, Changchun, China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qing-Hua Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Sen Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Cai-Rong Yang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xue-Qi Fu
- College of Life Sciences, Jilin University, Changchun, China
| | - Yi Hou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhao-Yi Wang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, Omaha, Nebraska, United States of America
| | - Jun Sheng
- College of Life Sciences, Jilin University, Changchun, China
- Yunnan Agricultural University, Kunming, China
- * E-mail: (JS); (Q-YS)
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (JS); (Q-YS)
| |
Collapse
|
10
|
Jeng YJ, Kochukov M, Watson CS. Combinations of physiologic estrogens with xenoestrogens alter calcium and kinase responses, prolactin release, and membrane estrogen receptor trafficking in rat pituitary cells. Environ Health 2010; 9:61. [PMID: 20950447 PMCID: PMC2967504 DOI: 10.1186/1476-069x-9-61] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 10/15/2010] [Indexed: 05/18/2023]
Abstract
BACKGROUND Xenoestrogens such as alkylphenols and the structurally related plastic byproduct bisphenol A have recently been shown to act potently via nongenomic signaling pathways and the membrane version of estrogen receptor-α. Though the responses to these compounds are typically measured individually, they usually contaminate organisms that already have endogenous estrogens present. Therefore, we used quantitative medium-throughput screening assays to measure the effects of physiologic estrogens in combination with these xenoestrogens. METHODS We studied the effects of low concentrations of endogenous estrogens (estradiol, estriol, and estrone) at 10 pM (representing pre-development levels), and 1 nM (representing higher cycle-dependent and pregnancy levels) in combinations with the same levels of xenoestrogens in GH3/B6/F10 pituitary cells. These levels of xenoestrogens represent extremely low contamination levels. We monitored calcium entry into cells using Fura-2 fluorescence imaging of single cells. Prolactin release was measured by radio-immunoassay. Extracellular-regulated kinase (1 and 2) phospho-activations and the levels of three estrogen receptors in the cell membrane (ERα, ERβ, and GPER) were measured using a quantitative plate immunoassay of fixed cells either permeabilized or nonpermeabilized (respectively). RESULTS All xenoestrogens caused responses at these concentrations, and had disruptive effects on the actions of physiologic estrogens. Xenoestrogens reduced the % of cells that responded to estradiol via calcium channel opening. They also inhibited the activation (phosphorylation) of extracellular-regulated kinases at some concentrations. They either inhibited or enhanced rapid prolactin release, depending upon concentration. These latter two dose-responses were nonmonotonic, a characteristic of nongenomic estrogenic responses. CONCLUSIONS Responses mediated by endogenous estrogens representing different life stages are vulnerable to very low concentrations of these structurally related xenoestrogens. Because of their non-classical dose-responses, they must be studied in detail to pinpoint effective concentrations and the directions of response changes.
Collapse
Affiliation(s)
- Yow-Jiun Jeng
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mikhail Kochukov
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cheryl S Watson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
11
|
Rangasamy V, Mishra R, Mehrotra S, Sondarva G, Ray RS, Rao A, Chatterjee M, Rana B, Rana A. Estrogen suppresses MLK3-mediated apoptosis sensitivity in ER+ breast cancer cells. Cancer Res 2010; 70:1731-40. [PMID: 20145118 PMCID: PMC2963191 DOI: 10.1158/0008-5472.can-09-3492] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Little knowledge exists about the mechanisms by which estrogen can impede chemotherapy-induced cell death of breast cancer cells. 17beta-Estradiol (E(2)) hinders cytotoxic drug-induced cell death in estrogen receptor-positive (ER(+)) breast cancer cells. We noted that the activity of the proapoptotic mixed lineage kinase 3 (MLK3) kinase was relatively higher in estrogen receptor-negative (ER(-)) breast tumors, suggesting that E(2) might inhibit MLK3 activity. The kinase activities of MLK3 and its downstream target, c-Jun NH(2)-terminal kinase, were rapidly inhibited by E(2) in ER(+) but not in ER(-) cells. Specific knockdown of AKT1/2 prevented MLK3 inhibition by E(2), indicating that AKT mediated this event. Furthermore, MLK3 inhibition by E(2) involved phosphorylation of MLK3 Ser(674) by AKT, attenuating the proapoptotic function of MLK3. We found that a pan-MLK inhibitor (CEP-11004) limited Taxol-induced cell death and that E(2) accentuated this limitation. Taken together, our findings indicate that E(2) inhibits the proapoptotic function of MLK3 as a mechanism to limit cytotoxic drug-induced death of ER(+) breast cancer cells.
Collapse
Affiliation(s)
- Velusamy Rangasamy
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Rajakishore Mishra
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Suneet Mehrotra
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Gautam Sondarva
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Rajarshi S. Ray
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Arundhati Rao
- Department of Pathology, Scott and White Hospital and Texas A & M Health Science Center, College of Medicine, Temple, Texas 76504
| | - Malay Chatterjee
- Division of Biochemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Basabi Rana
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
- Hines Veterans Affairs Medical Center, Hines, Illinois 60141
| | - Ajay Rana
- Department of Pharmacology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
- Hines Veterans Affairs Medical Center, Hines, Illinois 60141
| |
Collapse
|
12
|
Zárate S, Jaita G, Zaldivar V, Radl DB, Eijo G, Ferraris J, Pisera D, Seilicovich A. Estrogens exert a rapid apoptotic action in anterior pituitary cells. Am J Physiol Endocrinol Metab 2009; 296:E664-71. [PMID: 19158323 DOI: 10.1152/ajpendo.90785.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
It is now accepted that estrogens not only stimulate lactotrope proliferation but also sensitize anterior pituitary cells to proapoptotic stimuli. In addition to their classical mechanism of action through binding to intracellular estrogen receptors (ERs), there is increasing evidence that estrogens exert rapid actions mediated by cell membrane-localized ERs (mERs). In the present study, we examined the involvement of membrane-initiated steroid signaling in the proapoptotic action of estradiol in primary cultures of anterior pituitary cells from ovariectomized rats by using estren, a synthetic estrogen with no effect on classical transcription and a cell-impermeable 17beta-estradiol conjugate (E2-BSA). Both compounds induced cell death of anterior pituitary cells after 60 min of incubation as assessed by flow cytometry and the [3-(4,5-dimethylthiazol-2-yl)]-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Estren, E2, and E2-BSA induced apoptosis of lactotropes and somatotropes as evaluated by the deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay and immunodetection of prolactin (PRL) and growth hormone (GH). The proapoptotic effect of E2-BSA was abrogated by ICI-182,780, an antagonist of ERs. The expression of membrane-associated ERalpha was observed in PRL- and GH-bearing cells. Our results indicate that estradiol is able to exert a rapid apoptotic action in anterior pituitary cells, especially lactotropes and somatotropes, by a mechanism triggered by mERs. This mechanism could be involved in anterior pituitary cell turnover.
Collapse
Affiliation(s)
- S Zárate
- Instituto de Investigaciones en Reproducción, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Noel SD, Keen KL, Baumann DI, Filardo EJ, Terasawa E. Involvement of G protein-coupled receptor 30 (GPR30) in rapid action of estrogen in primate LHRH neurons. Mol Endocrinol 2009; 23:349-59. [PMID: 19131510 PMCID: PMC2654512 DOI: 10.1210/me.2008-0299] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 12/30/2008] [Indexed: 12/27/2022] Open
Abstract
Previously, we have reported that 17beta-estradiol (E(2)) induces an increase in firing activity of primate LH-releasing hormone (LHRH) neurons. The present study investigates whether E(2) alters LHRH release as well as the pattern of intracellular calcium ([Ca(2+)](i)) oscillations and whether G protein-coupled receptor 30 (GPR30) plays a role in mediating the rapid E(2) action in primate LHRH neurons. Results are summarized: 1) E(2), the nuclear membrane-impermeable estrogen, estrogen-dendrimer conjugate, and the plasma membrane-impermeable estrogen, E(2)-BSA conjugate, all stimulated LHRH release within 10 min of exposure; 2) whereas the estrogen receptor antagonist, ICI 182,780, did not block the E(2)-induced LHRH release, E(2) application to cells treated with pertussis toxin failed to induce LHRH release; 3) GPR30 mRNA was expressed in olfactory placode cultures, and GPR30 protein was expressed in a subset of LHRH neurons; 4) pertussis toxin treatment blocked the E(2)-induced increase in [Ca(2+)](i) oscillations; 5) knockdown of GPR30 in primate LHRH neurons by transfection with small interfering RNA (siRNA) for GPR30 completely abrogated the E(2)-induced changes in [Ca(2+)](i) oscillations, whereas transfection with control siRNA did not; 6) the estrogen-dendrimer conjugate-induced increase in [Ca(2+)](i) oscillations also did not occur in LHRH neurons transfected with GPR30 siRNA; and 7) G1, a GPR30 agonist, resulted in changes in [Ca(2+)](i) oscillations, similar to those observed with E(2). Collectively, E(2) induces a rapid excitatory effect on primate LHRH neurons, and this rapid action of E(2) appears to be mediated, in part, through GPR30.
Collapse
Affiliation(s)
- Sekoni D Noel
- Wisconsin National Primate Research Center, University of Wisconsin, 1223 Capitol Court, Madison, Wisconsin 53715-1299.
| | | | | | | | | |
Collapse
|
14
|
Bolli A, Galluzzo P, Ascenzi P, Del Pozzo G, Manco I, Vietri MT, Mita L, Altucci L, Mita DG, Marino M. Laccase treatment impairs bisphenol A-induced cancer cell proliferation affecting estrogen receptor alpha-dependent rapid signals. IUBMB Life 2008; 60:843-52. [PMID: 18767177 DOI: 10.1002/iub.130] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A wide variety of environmental contaminants exert estrogenic actions in wildlife, laboratory animals, and in human beings through binding to nuclear estrogen receptors (ERs). Here, the mechanism(s) of bisphenol A (BPA) to induce cell proliferation and the occurrence of its bioremediation by treatment with laccase are reported. BPA, highly present in natural world and considered as a model of environmental estrogen action complexity, promotes human cancer cell proliferation via ERalpha-dependent signal transduction pathways. Similar to 17beta-estradiol, BPA increases the phosphorylation of both extracellular regulated kinase and AKT. Specific inhibitors of these kinase completely block the BPA effect on cancer cell proliferation. Notably, high BPA concentrations (i.e., 0.1 and 1 mM) are cytotoxic even in ERalpha-devoid cancer cells, indicating that an ERalpha-independent mechanism participates to BPA-induced cytotoxicity. On the other hand, BPA oxidation by laccase impairs the binding of this environmental estrogen to ERalpha loosing at all ERalpha-dependent effect on cancer cell proliferation. Moreover, the laccase-catalyzed oxidation of BPA reduces the BPA cytotoxic effect. Thus, laccase appears to impair BPA action(s), representing an invaluable bioremediation enzyme.
Collapse
Affiliation(s)
- Alessandro Bolli
- Department of Biology, University Roma Tre, Viale Guglielmo Marconi 446, Roma, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Albanito L, Lappano R, Madeo A, Chimento A, Prossnitz ER, Cappello AR, Dolce V, Abonante S, Pezzi V, Maggiolini M. G-protein-coupled receptor 30 and estrogen receptor-alpha are involved in the proliferative effects induced by atrazine in ovarian cancer cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1648-55. [PMID: 19079715 PMCID: PMC2599758 DOI: 10.1289/ehp.11297] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 07/18/2008] [Indexed: 05/03/2023]
Abstract
BACKGROUND Atrazine, one of the most common pesticide contaminants, has been shown to up-regulate aromatase activity in certain estrogen-sensitive tumors without binding or activating the estrogen receptor (ER). Recent investigations have demonstrated that the orphan G-protein-coupled receptor 30 (GPR30), which is structurally unrelated to the ER, mediates rapid actions of 17beta-estradiol and environmental estrogens. OBJECTIVES Given the ability of atrazine to exert estrogen-like activity in cancer cells, we evaluated the potential of atrazine to signal through GPR30 in stimulating biological responses in cancer cells. METHODS AND RESULTS Atrazine did not transactivate the endogenous ERalpha in different cancer cell contexts or chimeric proteins encoding the ERalpha and ERbeta hormone-binding domain in gene reporter assays. Moreover, atrazine neither regulated the expression of ERalpha nor stimulated aromatase activity. Interestingly, atrazine induced extracellular signal-regulated kinase (ERK) phosphorylation and the expression of estrogen target genes. Using specific signaling inhibitors and gene silencing, we demonstrated that atrazine stimulated the proliferation of ovarian cancer cells through the GPR30-epidermal growth factor receptor transduction pathway and the involvement of ERalpha. CONCLUSIONS Our results indicate a novel mechanism through which atrazine may exert relevant biological effects in cancer cells. On the basis of the present data, atrazine should be included among the environmental contaminants potentially able to signal via GPR30 in eliciting estrogenic action.
Collapse
Affiliation(s)
- Lidia Albanito
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Rosamaria Lappano
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Antonio Madeo
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Adele Chimento
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Eric R. Prossnitz
- Department of Cell Biology and Physiology and Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Vincenza Dolce
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Sergio Abonante
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | - Vincenzo Pezzi
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | | |
Collapse
|
16
|
Estradiol-induced enhancement of object memory consolidation involves hippocampal extracellular signal-regulated kinase activation and membrane-bound estrogen receptors. J Neurosci 2008; 28:8660-7. [PMID: 18753366 DOI: 10.1523/jneurosci.1968-08.2008] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The extracellular signal-regulated kinase (ERK) pathway is critical for various forms of learning and memory, and is activated by the potent estrogen 17beta-estradiol (E(2)). Here, we asked whether E(2) modulates memory via ERK activation and putative membrane-bound estrogen receptors (ERs). Using ovariectomized mice, we first demonstrate that intraperitoneal injection of 0.2 mg/kg E(2) significantly increases dorsal hippocampal levels of phosphorylated ERK protein 1 h after injection. Second, we show that E(2) administered intraperitoneally (0.2 mg/kg) or via intrahippocampal infusion (5.0 microg/side) immediately after training in an object recognition task significantly enhances memory retention, and that the beneficial effect of intraperitoneal E(2) is blocked by dorsal hippocampal inhibition of ERK activation. Third, using bovine serum albumin-conjugated 17beta-estradiol (BSA-E(2)), we demonstrate that E(2) binding at membrane-bound ERs can increase dorsal hippocampal ERK activation and enhance object memory consolidation in an ERK-dependent manner. Fourth, we show that this effect is independent of nuclear ERs, but is dependent on the dorsal hippocampus. By demonstrating that E(2) enhances memory consolidation via dorsal hippocampal ERK activation, this study is the first to identify a specific molecular pathway by which E(2) modulates memory and to demonstrate a novel role for membrane-bound ERs in mediating E(2)-induced improvements in hippocampal memory consolidation.
Collapse
|
17
|
Wang Y, Zhao Y, Chen X. Experimental study on the estrogen-like effect of boric Acid. Biol Trace Elem Res 2008; 121:160-70. [PMID: 17943231 DOI: 10.1007/s12011-007-8041-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 10/01/2007] [Indexed: 11/24/2022]
Abstract
There are now considerable evidences that boric acid has reproductive and developmental toxicity, but it is uncertain whether such toxicity is caused by estrogen-like effect. Our objective is to determine the estrogen-like effect of boric acid. Proliferation assay of MCF-7 human breast cancer cells, uterotrophic assay, measure assay of the estradiol (E2), proliferation assay of mucous membrane cells, and assay of estrogen receptor were conducted in this study. Boric acid could increase the weight of uterus of ovariectomized SD rats and the height of epithelium cells of mucous membrane, enhance the expression of the proliferating cell nucleus antigen, and reduce the density of estrogen receptors. However, boric acid could not affect the level of estradiol in serum and stimulate the proliferation of MCF-7 human breast cancer cells. In this study, boric acid exhibited the estrogen-like effect in vivo.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Zhengzhou University of Henan, No. 100 Science Road, 450001 Zhengzhou, People's Republic of China
| | | | | |
Collapse
|
18
|
González M, Reyes R, Damas C, Alonso R, Bello AR. Oestrogen receptor alpha and beta in female rat pituitary cells: an immunochemical study. Gen Comp Endocrinol 2008; 155:857-68. [PMID: 18067893 DOI: 10.1016/j.ygcen.2007.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 10/17/2007] [Accepted: 10/23/2007] [Indexed: 01/05/2023]
Abstract
Estradiol is a critical factor in the anterior pituitary secretory activity of mammalian females. Previous reports have demonstrated the presence of oestrogen receptor alpha (ERalpha) and beta (ERbeta) in specific anterior pituitary cells from ovariectomized rats, as well as in the whole anterior pituitary at particular stages of the rat oestrous cycle. However, the ERalpha and ERbeta distribution patterns in specific hormone producing cells of the anterior pituitary during the oestrous cycle remain to be clarified. The purpose of this study was to determine the cellular and subcellular distribution of both ER-subtypes during the rat oestrous cycle, using immunochemistry at light- and electron-microscope levels. ERalpha-immunoreactive (ir) cells mainly corresponded to PRL-ir cells and, to a lesser extent, to TSH-, FSH- and GH-ir cells. ERbeta-ir cells corresponded to a few GH-, PRL- and FSH-ir cells, whichever the phase of the cycle. ERalpha-ir was found either in the cytoplasm and/or the nucleus, depending on the phase of the oestrous cycle, while ERbeta-ir was always detected in the cytoplasm. Both ER-subtypes were immunoreactives inside the rough endoplasmic reticulum (RER), secretory vesicles (SV) and free in the cytosol. The highest number of ERalpha-ir cells was consistently found at pro-oestrus midday and the lowest at metaoestrous, while the number of ERbeta-ir cells was low in all stages of the cycle. These results indicate that the genomic actions of oestrogen in the anterior pituitary cells during the oestrous cycle are mediated by ERalpha. However, the localization of ERalpha and ERbeta in the RER and SV suggest a different translational and/or post-translational pathway, which could be involved in non-genomic mechanisms.
Collapse
Affiliation(s)
- Miriam González
- Cell Biology Section, University of La Laguna School of Biology and FICIC, 38230 La Laguna, Tenerife, Spain
| | | | | | | | | |
Collapse
|
19
|
Santen RJ, Song RX, Masamura S, Yue W, Fan P, Sogon T, Hayashi SI, Nakachi K, Eguchi H. Adaptation to Estradiol Deprivation Causes Up-Regulation of Growth Factor Pathways and Hypersensitivity to Estradiol in Breast Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 630:19-34. [DOI: 10.1007/978-0-387-78818-0_2] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
20
|
Boulware MI, Kordasiewicz H, Mermelstein PG. Caveolin proteins are essential for distinct effects of membrane estrogen receptors in neurons. J Neurosci 2007; 27:9941-50. [PMID: 17855608 PMCID: PMC6672640 DOI: 10.1523/jneurosci.1647-07.2007] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
It has become widely accepted that along with its ability to directly regulate gene expression, estradiol also influences cell signaling and brain function via rapid membrane-initiated events. Many of these novel signaling processes are dependent on estrogen receptors (ERs) localized to the neuronal membrane. However, the mechanism(s) by which ERs are able to trigger cell signaling when targeted to the neuronal membrane surface has yet to be determined. In hippocampal neurons, we find that caveolin proteins are essential for the regulation of CREB (cAMP response element-binding protein) phosphorylation after estradiol activation of metabotropic glutamate receptor (mGluR) signaling. Furthermore, caveolin-1 (CAV1) and CAV3 differentially regulate the ability of estradiol to activate two discrete signaling pathways. ER alpha activation of mGluR1a is dependent on CAV1, whereas CAV3 is necessary for ER alpha and ER beta activation of mGluR2/3. These results are consistent with previous reports in non-neuronal cells, implicating the importance of caveolin proteins in rapid estrogen signaling. In addition, the functional isolation of distinct estrogen-sensitive signaling pathways by different caveolin proteins suggests novel mechanisms through which the membrane-initiated effects of estradiol are orchestrated.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Holly Kordasiewicz
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
21
|
Zhang X, Wang Y, Zhao Y, Chen X. Experimental study on the estrogen-like effect of mercuric chloride. Biometals 2007; 21:143-50. [PMID: 17588195 DOI: 10.1007/s10534-007-9102-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Accepted: 05/23/2007] [Indexed: 10/23/2022]
Abstract
Although mercuric chloride has toxicity on reproductive system, it is uncertain if such toxicity is induced by estrogen-like effect. To study whether mercuric chloride has the estrogen-like effect and its relevant mechanism, proliferation assay of MCF-7 human breast cancer cells, uterotrophic assay, peroxidase activity assay and estrogen receptor competitive binding assay were conducted to screen the estrogen-like effect of mercuric chloride. The MCF-7 cells proliferated in the stimulation of mercuric chloride and got to the peak at 10(-7) mol/l concentration. And this proliferation could be completely blocked by estrogenic antagonist ICI182.780. In addition, mercuric chloride could increase the weight of uterus of ovariectomized SD rats and the peroxidase activity of uterus complying with dose-effect relationship. However, mercuric chloride could not affect the binding of estradiol (E(2)) to estrogen receptor (ER). So mercuric chloride exhibits the estrogen-like effect through binding and activating ER rather than bind to ER by competing with E(2).
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Toxicology, Zhengzhou University of Henan, Zhengzhou, PR China
| | | | | | | |
Collapse
|
22
|
Roy D, Cai Q, Felty Q, Narayan S. Estrogen-induced generation of reactive oxygen and nitrogen species, gene damage, and estrogen-dependent cancers. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2007; 10:235-57. [PMID: 17620201 DOI: 10.1080/15287390600974924] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In addition to the direct effect of estrogen on mitochondria and the redox cycling of catechol estrogen, estrogen-induced proinflammatory cytokines, such as interleukin-1 beta (IL-1beta) and tumor necrosis factor alpha (TNF-alpha), also generate reactive oxygen and nitrogen species (RO/NS). Different cellular signaling pathways may operate in response to varying levels of estrogen-induced RO/NS, leading to genotoxic damage, cell apoptosis, or cell growth. At high levels of RO/NS, cells receiving genotoxic insults, if not repaired, may engage the apoptotic pathways. There is increasing evidence supporting that estrogen-induced alterations in the genome of cells is produced by oxidative attack. Furthermore, ROS generated by estrogen exposure and/or active metabolites of estrogen in combination with receptor-mediated proliferation of genetically damaged cells may be involved in tumor development. This view is supported by the findings of DNA modifications produced in vitro or in vivo by natural and synthetic estrogens in the target organs of cancer both in experimental models and in humans. Interaction of estrogen-induced oxidants and estrogen metabolites with DNA was shown to generate mutations in genes. Cotreatment with an inhibitor of IL-1beta and TNF-alpha synthesis, pentoxifylline, decreased stilbene estrogen-induced levels of myeloperoxidase (MPO), 8-hydroxydeoxyguanosine formation, and gene mutations, and prevented stilbene estrogen-induced lesions. Stable MCF-7 clones overexpressing IL-1beta resulted in a high level of IL-1beta peptide secretion undergoing cell apoptosis, and an elevated level of p53 protein in response to high oxidative stress when compared to nontransfected cells, whereas MCF-7 clones overexpressing IL-1beta that resulted in a moderate level of IL-1beta secretion stimulated the clonal expansion of MCF-7 and TM3 cells. Estrogen-induced MCF-7 cell growth and cyclin D1 expression were suppressed by antioxidants and mitochondrial blockers. These studies support that in addition to ovarian estrogen-mediated ER signaling, mitogenic signals may also come from estrogen-induced RO/NS. Further validation of this concept that the concentration of the RO/NS within the cellular microenvironment determines its stimulatory or inhibitory growth signals as well as its genotoxic effects regulating the growth of estrogen-dependent tumors may result in novel preventive strategies.
Collapse
Affiliation(s)
- Deodutta Roy
- Department of Environmental and Occupational Health, Florida International University, Miami, Florida 33199, USA.
| | | | | | | |
Collapse
|
23
|
Wetherill YB, Akingbemi BT, Kanno J, McLachlan JA, Nadal A, Sonnenschein C, Watson CS, Zoeller RT, Belcher SM. In vitro molecular mechanisms of bisphenol A action. Reprod Toxicol 2007; 24:178-98. [PMID: 17628395 DOI: 10.1016/j.reprotox.2007.05.010] [Citation(s) in RCA: 640] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Accepted: 05/18/2007] [Indexed: 11/29/2022]
Abstract
Bisphenol A (BPA, 2,2-bis (4-hydroxyphenyl) propane; CAS# 80-05-7) is a chemical used primarily in the manufacture of polycarbonate plastic, epoxy resins and as a non-polymer additive to other plastics. Recent evidence has demonstrated that human and wildlife populations are exposed to levels of BPA which cause adverse reproductive and developmental effects in a number of different wildlife species and laboratory animal models. However, there are major uncertainties surrounding the spectrum of BPA's mechanisms of action, the tissue-specific impacts of exposures, and the critical windows of susceptibility during which target tissues are sensitive to BPA exposures. As a foundation to address some of those uncertainties, this review was prepared by the "In vitro" expert sub-panel assembled during the "Bisphenol A: An Examination of the Relevance of Ecological, In vitro and Laboratory Animal Studies for Assessing Risks to Human Health" workshop held in Chapel Hill, NC, Nov 28-29, 2006. The specific charge of this expert panel was to review and assess the strength of the published literature pertaining to the mechanisms of BPA action. The resulting document is a detailed review of published studies that have focused on the mechanistic basis of BPA action in diverse experimental models and an assessment of the strength of the evidence regarding the published BPA research.
Collapse
Affiliation(s)
- Yelena B Wetherill
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Narita SI, Goldblum RM, Watson CS, Brooks EG, Estes DM, Curran EM, Midoro-Horiuti T. Environmental estrogens induce mast cell degranulation and enhance IgE-mediated release of allergic mediators. ENVIRONMENTAL HEALTH PERSPECTIVES 2007; 115:48-52. [PMID: 17366818 PMCID: PMC1797832 DOI: 10.1289/ehp.9378] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
BACKGROUND Prevalence and morbidity of allergic diseases have increased over the last decades. Based on the recently recognized differences in asthma prevalence between the sexes, we have examined the effect of endogenous estrogens on a key element of the allergic response. Some lipophilic pollutants have estrogen-like activities and are termed environmental estrogens. These pollutants tend to degrade slowly in the environment and to bioaccumulate and bioconcentrate in the food chain; they also have long biological half-lives. OBJECTIVES Our goal in this study was to identify possible pathogenic roles for environmental estrogens in the development of allergic diseases. METHODS We screened a number of environmental estrogens for their ability to modulate the release of allergic mediators from mast cells. We incubated a human mast cell line and primary mast cell cultures derived from bone marrow of wild type and estrogen receptor alpha (ER-alpha)-deficient mice with environmental estrogens with and without estradiol or IgE and allergens. We assessed degranulation of mast cells by quantifying the release of beta-hexosaminidase. RESULTS All of the environmental estrogens tested caused rapid, dose-related release of beta-hexosaminidase from mast cells and enhanced IgE-mediated release. The combination of physiologic concentrations of 17beta-estradiol and several concentrations of environmental estrogens had additive effects on mast cell degranulation. Comparison of bone marrow mast cells from ER-alpha-sufficient and ER-alpha-deficient mice indicated that much of the effect of environmental estrogens was mediated by ER-alpha. CONCLUSIONS Our findings suggest that estrogenic environmental pollutants might promote allergic diseases by inducing and enhancing mast cell degranulation by physiologic estrogens and exposure to allergens.
Collapse
Affiliation(s)
| | | | - Cheryl S. Watson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - D. Mark Estes
- Department of Pediatrics, Child Health Research Center and
| | | | - Terumi Midoro-Horiuti
- Department of Pediatrics, Child Health Research Center and
- Address correspondence to T. Midoro-Horiuti, Child Health Research Center, University of Texas Medical Branch, 2.300 Children’s Hospital, 301 University Blvd., Galveston, TX 77555-0366 USA. Telephone: (409) 772-3832. Fax: (409) 772-1761. E-mail:
| |
Collapse
|
25
|
Hayashi H, Nishimoto A, Oshima N, Iwamuro S. EXPRESSION OF THE ESTROGEN RECEPTOR ALPHA GENE IN THE ANAL FIN OF JAPANESE MEDAKA, ORYZIAS LATIPES, BY ENVIRONMENTAL CONCENTRATIONS OF BISPHENOL A. J Toxicol Sci 2007; 32:91-6. [PMID: 17327697 DOI: 10.2131/jts.32.91] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The anal fin in Japanese medaka, Oryzias latipes, is a typical sexual secondary character. In the present study, we focused on this organ and examined the effects of low doses of a natural estrogen, 17beta-estradiol (E(2)), and an environmental xenoestrogen, bisphenol A (BPA), in vivo by monitoring estrogen receptor (ER) alpha gene expression. Groups of adult male and female medaka were immersed in 10(-9) M E(2) or 10(-10) to 10(-8) M BPA and the levels of ERalpha gene transcripts in the anal fins were measured by semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). One day of treatment with each concentration of BPA examined and 10(-9) M E(2) increased the levels of ERalpha mRNA in female anal fins by 3-fold as compared with controls. In the male specimens, neither 10(-9) M E(2) nor 10(-10) M BPA showed remarkable effects on the anal fins as compared with the results in females, but 10(-9) and 10(-8) M BPA increased the levels of ERalpha mRNA by 2.3- and 3.3-fold with 1 day of exposure, respectively. The present results showed that medaka anal fins may be a sensitive bio-indicator for screening of environmental estrogenic chemicals.
Collapse
|
26
|
Abstract
By regulating activities and expression levels of key signaling molecules, estrogens control mechanisms that are responsible for crucial cellular functions. Ligand binding to estrogen receptor (ER) leads to conformational changes that regulate the receptor activity, its interaction with other proteins and DNA. In the cytoplasm, receptor interactions with kinases and scaffolding molecules regulate cell signaling cascades (extranuclear/nongenomic action). In the nucleus, estrogens control a repertoire of coregulators and other auxiliary proteins that are associated with ER, which in turn determines the nature of regulated genes and level of their expression (genomic action). The combination of genomic and nongenomic actions of estrogens ultimately confers the cell-type and tissue-type selectivity. Recent studies have revealed some important new insights into the molecular mechanisms underlying ER action, which may help to explain the functional basis of existing selective ER modulators (SERMs) and provide evidence into how ER might be selectively targeted to achieve specific therapeutic goals. In this review, we will summarize some new molecular details that relate to estrogen signaling. We will also discuss some new strategies that may potentially lead to the development of functionally selective ER modulators that can separate between the beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS as well as the "detrimental," proliferative effects in reproductive tissues and organs.
Collapse
Affiliation(s)
- Boris J Cheskis
- Women's Health and Musculoskeletal Biology, Wyeth Research, Collegeville, Pennsylvania 19426, USA.
| | | | | | | |
Collapse
|
27
|
Xenoestrogens are potent activators of nongenomic estrogenic responses. Steroids 2006; 72:124-34. [PMID: 17174995 DOI: 10.1016/j.steroids.2006.11.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 10/31/2006] [Accepted: 11/03/2006] [Indexed: 01/28/2023]
Abstract
Studies of the nuclear transcriptional regulatory activities of non-physiological estrogens have not explained their actions in mediating endocrine disruption in animals and humans at the low concentrations widespread in the environment. However, xenoestrogens have rarely been tested for their ability to participate in the plethora of nongenomic steroid signaling pathways elucidated over the last several years. Here we review what is known about such responses in comparison to our recent evidence that xenoestrogens can rapidly and potently elicit signaling through nongenomic pathways culminating in functional endpoints. Both estradiol (E(2)) and compounds representing various classes of xenoestrogens (diethylstilbestrol, coumestrol, bisphenol A, DDE, nonylphenol, endosulfan, and dieldrin) act via a membrane version of the estrogen receptor-alpha on pituitary cells, and can provoke Ca(2+) influx via L-type channels, leading to prolactin (PRL) secretion. These hormones and mimetics can also cause the oscillating activation of extracellular regulated kinases (ERKs). However, individual estrogen mimetics differ in their potency and temporal phasing of these activations compared to each other and to E(2). It is perhaps in these ways that they disrupt some endocrine functions when acting in combination with physiological estrogens. Our quantitative assays allow comparison of these outcomes for each mimetic, and let us build a detailed picture of alternative signaling pathway usage. Such an understanding should allow us to determine the estrogenic or antiestrogenic potential of different types of xenoestrogens, and help us to develop strategies for preventing xenoestrogenic disruption of estrogen action in many tissues.
Collapse
|
28
|
Thomas P, Dong J. Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: a potential novel mechanism of endocrine disruption. J Steroid Biochem Mol Biol 2006; 102:175-9. [PMID: 17088055 DOI: 10.1016/j.jsbmb.2006.09.017] [Citation(s) in RCA: 424] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A wide variety of environmental contaminants have been shown to exert estrogenic actions in wildlife and laboratory animals through binding to nuclear estrogen receptors (ERs) and subsequent transcription of estrogen responsive genes. We show here that several of these environmental estrogens also bind to the novel seven-transmembrane estrogen receptor, GPR30, to activate alternative estrogen signaling pathways in an ER-negative cell line (HEK293) stably transfected with the receptor. Genestein was the most effective competitor for the receptor (IC(50) 133 nM), with a relative binding affinity (RBA) 13% that of estradiol-17beta (E2). Bisphenol A, zearalonone, and nonylphenol also had relatively high binding affinities for GPR30 with RBAs of 2-3%. Kepone, p,p'-DDT, 2,2',5',-PCB-4-OH and o,p'-DDE had lower affinities with RBAs of 0.25-1.3%, whereas o,p'-DDT, p,p'-DDE, methoxychlor and atrazine caused less than 50% displacement of [(3)H]-E2 at concentrations up to 10 microM. Overall, the binding affinities of these compounds for GPR30 are broadly similar to their affinities to the ERs. Environmental estrogens with relatively high binding affinities for GPR30 (genestein, bisphenol A, nonylphenol and Kepone) also displayed estrogen agonist activities in an in vitro assay of membrane-bound adenylyl cyclase activity, a GPR30-dependent signaling pathway activated by estrogens. The results indicate that nontraditional estrogen actions mediated through GPR30 are potentially susceptible to disruption by a variety of environmental estrogens.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| | | |
Collapse
|
29
|
Kansra S, Yamagata S, Sneade L, Foster L, Ben-Jonathan N. Differential effects of estrogen receptor antagonists on pituitary lactotroph proliferation and prolactin release. Mol Cell Endocrinol 2005; 239:27-36. [PMID: 15950373 DOI: 10.1016/j.mce.2005.04.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Revised: 04/22/2005] [Accepted: 04/25/2005] [Indexed: 11/25/2022]
Abstract
Anti-estrogens act by inhibiting estrogen receptor (ER) function. Unlike raloxifene and tamoxifen which exhibit both antagonist and agonist properties, ICI 182,780 (ICI) is considered a "pure" anti-estrogen devoid of any agonistic activities. Whereas there is ample information on the effects of anti-estrogens on the breast and uterus, little is known about their action on the pituitary, the estrogen-sensitive master endocrine gland. Our objectives were to: (1) compare the effects of ICI, tamoxifen and raloxifene on lactotroph proliferation in the absence of estrogen, (2) determine whether their action is mediated through the ER, and (3) compare their effects on prolactin (PRL) release. We are reporting that ICI is a potent inhibitor of lactotroph proliferation (both GH3 and MMQ cells) with maximal inhibition of 45-50% seen with 1nM. ICI is several orders of magnitude more potent than raloxifene while tamoxifen has no effect. Neither anti-estrogen affects T47D breast cancer cell proliferation. GH3 cell incubation with ICI for 1h only causes maximal suppression of cell proliferation, an effect which is reversed by co-incubation with estrogen. Such a short exposure to ICI is sufficient to cause rapid and persistent downregulation of ERalpha protein, whereas downregulation of ERbeta is significantly delayed; tamoxifen and raloxifene have no appreciable effects on ER(s) levels. The ability of ICI to inhibit GH3 cell proliferation is dependent upon ERalpha, since an ERalpha, but not ERbeta, specific agonist reverses the effect of ICI. PRL release is differentially regulated by the anti-estrogens. ICI at 0.1nM suppresses PRL release from GH3 cells by 80%, with a similar strong suppression also seen with 10nM raloxifene. However, tamoxifen at 0.01nM inhibits PRL release but has no effect at 10nM. Cell co-incubation with ICI and estradiol results in a four-fold increase in PRL release. Taken together, our study shows that ICI, in the absence of exogenous estrogens, inhibits lactotroph proliferation and PRL release by downregulating or inactivating ERalpha. The dissimilar responses of cell proliferation and PRL release to the anti-estrogens suggest that both processes are regulated by different mechanisms. These data highlight the importance of studying the effects of anti-estrogens in multiple systems.
Collapse
Affiliation(s)
- Sanjay Kansra
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA.
| | | | | | | | | |
Collapse
|
30
|
Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein PG. Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci 2005; 25:5066-78. [PMID: 15901789 PMCID: PMC6724851 DOI: 10.1523/jneurosci.1427-05.2005] [Citation(s) in RCA: 315] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Revised: 04/13/2005] [Accepted: 04/16/2005] [Indexed: 12/12/2022] Open
Abstract
In addition to mediating sexual maturation and reproduction through stimulation of classical intracellular receptors that bind DNA and regulate gene expression, estradiol is also thought to influence various brain functions by acting on receptors localized to the neuronal membrane surface. Many intracellular signaling pathways and modulatory proteins are affected by estradiol via this unconventional route, including regulation of the transcription factor cAMP response element-binding protein (CREB). However, the mechanisms by which estradiol acts at the membrane surface are poorly understood. Because both estradiol and CREB have been implicated in regulating learning and memory, we characterized the effects of estradiol on this transcription factor in cultured rat hippocampal neurons. Within minutes of administration, estradiol triggered mitogen-activated protein kinase (MAPK)-dependent CREB phosphorylation in unstimulated neurons. Furthermore, after brief depolarization, estradiol attenuated L-type calcium channel-mediated CREB phosphorylation. Thus, estradiol exhibited both positive and negative influences on CREB activity. These effects of estradiol were sex specific and traced to membrane-localized estrogen receptors that stimulated group I and II metabotropic glutamate receptor (mGluR) signaling. Activation of estrogen receptor alpha (ERalpha) led to mGluR1a signaling, triggering CREB phosphorylation through phospholipase C regulation of MAPK. In addition, estradiol stimulation of ERalpha or ERbeta triggered mGluR2/3 signaling, decreasing L-type calcium channel-mediated CREB phosphorylation. These results not only characterize estradiol regulation of CREB but also provide two putative signaling mechanisms that may account for many of the unexplained observations regarding the influence of estradiol on nervous system function.
Collapse
Affiliation(s)
- Marissa I Boulware
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The female sex steroid hormones 17beta-estradiol and progesterone mediate their biological effects on development, differentiation, and maintenance of reproductive tract and other target tissues through gene regulation by nuclear steroid receptors that function as ligand-dependent transcription factors. However, not all effects of 17beta-estradiol and progesterone are mediated by direct control of gene expression. These hormones also have rapid stimulatory effects on the activities of a variety of signal transduction molecules and pathways and, in many cases, these effects appear to be initiated from the plasma cell membrane. There is growing evidence that a subpopulation of the conventional nuclear steroid receptor localized at the cell membrane mediates many of the rapid signaling actions of steroid hormones; however, novel membrane receptors unrelated to conventional steroid receptors have also been implicated. This chapter reviews the nature of the receptors that mediate rapid signaling actions of estrogen and progesterone and describes the signaling molecules and pathways involved, the mechanisms by which receptors couple with components of signaling complexes and trigger responses, and the target tissues and cell functions regulated by this mode of steroid hormone action.
Collapse
Affiliation(s)
- Dean P Edwards
- University of Colorado Health Sciences Center, Department of Pathology and Program in Molecular Biology, Aurora, Colorado 80045, USA.
| |
Collapse
|
32
|
Santen RJ, Song RX, Zhang Z, Kumar R, Jeng MH, Masamura S, Lawrence J, MacMahon LP, Yue W, Berstein L. Adaptive hypersensitivity to estrogen: mechanisms and clinical relevance to aromatase inhibitor therapy in breast cancer treatment. J Steroid Biochem Mol Biol 2005; 95:155-65. [PMID: 16024245 DOI: 10.1016/j.jsbmb.2005.04.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Breast tumors in women can adapt to endocrine deprivation therapy by developing hypersensitivity to estradiol. For this reason, aromatase inhibitors can be effective in women relapsing after treatment with tamoxifen or following oophorectomy. To understand the mechanisms responsible, we examined estrogenic stimulation of cell proliferation in a model system and provided in vitro and in vivo evidence that long-term estradiol deprivation (LTED) causes "adaptive hypersensitivity". The primary mechanisms responsible involve up-regulation of ER alpha as well as the MAP kinase, PI-3 kinase, and mTOR growth factor pathways. ER alpha is 4-10-fold up-regulated and co-opts a classical growth factor pathway using Shc, Grb2, and Sos. This induces rapid non-genomic effects which are enhanced in LTED cells. Estradiol binds to cell membrane associated ER alpha, physically associates with the adaptor protein Shc, and induces its phosphorylation. In turn, Shc binds Grb2 and Sos which result in the rapid activation of MAP kinase. These non-genomic effects of estradiol produce biologic effects as evidenced by Elk activation and by morphologic changes in cell membranes. Additional effects include activation of PI-3 kinase and mTOR pathways through estradiol induced binding of ER alpha to the IGF-1 and EGF receptors. Further proof of the non-genomic effects of estradiol involved use of "designer" cells which selectively express ER alpha in nucleus, cytosol, and cell membrane. We have used a new downstream inhibitor of these pathways, farnesyl-thio-salicylic acid (FTS), to block proliferation in hypersensitive cells as a model for a potentially effective strategy for treatment of patients.
Collapse
Affiliation(s)
- R J Santen
- Department of Medicine, University of Virginia Health System, P.O. Box 801416, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wozniak AL, Bulayeva NN, Watson CS. Xenoestrogens at picomolar to nanomolar concentrations trigger membrane estrogen receptor-alpha-mediated Ca2+ fluxes and prolactin release in GH3/B6 pituitary tumor cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2005; 113:431-9. [PMID: 15811834 PMCID: PMC1278483 DOI: 10.1289/ehp.7505] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Xenoestrogens (XEs) are widespread in our environment and are known to have deleterious effects in animal (and perhaps human) populations. Acting as inappropriate estrogens, XEs are thought to interfere with endogenous estrogens such as estradiol (E2) to disrupt normal estrogenic signaling. We investigated the effects of E2 versus several XEs representing organochlorine pesticides (dieldrin, endosulfan, o',p'-dichlorodiphenylethylene), plastics manufacturing by-products/detergents (nonylphenol, bisphenol A), a phytoestrogen (coumestrol), and a synthetic estrogen (diethylstilbestrol) on the pituitary tumor cell subline GH3/B6/F10, previously selected for expression of high levels of membrane estrogen receptor-alpha. Picomolar to nanomolar concentrations of both E2 and XEs caused intracellular Ca2+ changes within 30 sec of administration. Each XE produced a unique temporal pattern of Ca2+ elevation. Removing Ca2+ from the extracellular solution abolished both spontaneous and XE-induced intracellular Ca2+ changes, as did 10 microM nifedipine. This suggests that XEs mediate their actions via voltage-dependent L-type Ca2+ channels in the plasma membrane. None of the Ca2+ fluxes came from intracellular Ca2+ stores. E2 and each XE also caused unique time- and concentration-dependent patterns of prolactin (PRL) secretion that were largely complete within 3 min of administration. PRL secretion was also blocked by nifedipine, demonstrating a correlation between Ca2+ influx and PRL secretion. These data indicate that at very low concentrations, XEs mediate membrane-initiated intracellular CCa2+ increases resulting in PRL secretion via a mechanism similar to that for E2, but with distinct patterns and potencies that could explain their abilities to disrupt endocrine functions.
Collapse
Affiliation(s)
- Ann L Wozniak
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston, Texas 77555-0645, USA
| | | | | |
Collapse
|
34
|
Walsh DE, Dockery P, Doolan CM. Estrogen receptor independent rapid non-genomic effects of environmental estrogens on [Ca2+]i in human breast cancer cells. Mol Cell Endocrinol 2005; 230:23-30. [PMID: 15664448 DOI: 10.1016/j.mce.2004.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Revised: 11/12/2004] [Accepted: 11/15/2004] [Indexed: 11/27/2022]
Abstract
The aim of this study was to identify and characterize an alternative pathway through which environmental estrogenic compounds may mediate their intracellular effects. Three human breast cancer cell lines were employed including MCF-7 cells, which express both ERalpha and ERbeta; MDA-MB-231 cells, which express ERbeta but not ERalpha; and SKBR-3 cells, which express neither ERalpha nor ERbeta. The effect of environmental estrogenic compounds on intracellular calcium ion concentration ([Ca(2+)](i)) was measured and compared to that of 17beta-estradiol (E2). A rapid and maintained increase in [Ca(2+)](i) was observed following the application of nanomolar concentrations of environmental estrogens and E2 regardless of the expression of ERalpha and ERbeta. Removal of extracellular Ca(2+) completely abolished the steroid-induced [Ca(2+)](i) increase. Pre-treatment of cells with the estrogen receptor (ER) antagonist ICI 182,780 had no effect on either basal [Ca(2+)](i) or the steroid-triggered [Ca(2+)](i) response. In summary, we have demonstrated ER independent rapid non-genomic effects of environmental estrogenic compounds, at nanomolar concentrations, on [Ca(2+)](i). The results of this study demonstrate an alternative pathway to explain potent intracellular effects of endocrine disrupting chemicals.
Collapse
Affiliation(s)
- Deirdre E Walsh
- Department of Physiology, Biosciences Institute, University College Cork, Cork, Ireland.
| | | | | |
Collapse
|
35
|
Abstract
Estrogen has multifaceted effects on the hypothalamus that regulate a number of homeostatic functions including reproduction, temperature, energy balance, stress, and motivated behaviors. Estrogen targets all of the major hypothalamic neuroendocrine and autonomic cellular groups to activate multiple signaling pathways. Originally it was thought that all of these actions of estrogen could be ascribed to its binding to its "classical" intracellular receptor and to alterations in gene transcription. However, we now know that this steroid hormone activates multiple signaling pathways to affect neuronal excitability and gene transcription. Although the "classical" genomic signaling pathway has been recognized for almost half a century, until recently little attention has been paid to the rapid membrane-initiated signaling by estrogen in neurons. It has been known since the 1970s that estrogen can rapidly alter neuronal firing within seconds, indicating that some cellular effects of estrogen could occur via rapid, non-transcriptional mechanisms. Therefore, this chapter reviews the current status of estrogen signaling in the hypothalamus via membrane-initiated and nuclear-mediated events that affect the excitability of hypothalamic neurons and, ultimately, neuroendocrine and autonomic functions.
Collapse
Affiliation(s)
- Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University Portland, Oregon 97239, USA
| | | | | |
Collapse
|
36
|
Thomas P, Doughty K. Disruption of rapid, nongenomic steroid actions by environmental chemicals: interference with progestin stimulation of sperm motility in Atlantic croaker. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2004; 38:6328-6332. [PMID: 15597889 DOI: 10.1021/es0403662] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Several nongenomic steroid actions, like genomic ones, can be disrupted by estrogenic xenobiotics (xenoestrogens), but the extent and sensitivity of this alternative mechanism of steroid action to chemical interference remain unclear. The effects of environmentally realistic concentrations of a broad range of organic contaminants on the nongenomic action of a progestin (17,20beta,21-trihydroxy-4-pregnen-3-one or 20beta-S) to upregulate Atlantic croaker sperm motility were examined in an in vitro bioassay. Pretreatment of sperm for 10 min in vitro with estrogenic compounds (estradiol-17beta, o,p'-DDT derivatives, zearalenone, bisphenol A, 2',3',4',5'-PCB-4-OH, kepone, chlordane, methoxyclor) and nonestrogenic organic compounds (p,p'-DDT derivatives, atrazine, Aroclor 1254, naphthalene, benzene) at concentrations ranging from 0.01 to 10 microM did not decrease the percent of motile sperm, but all of them partially or completely blocked the response to 20beta-S. Most of the compounds impaired this endocrine mechanism at a concentration of 0.1 microM (approximately 30-40ppb), whereas o,p'-DDT and atrazine were effective at lower concentrations. The antagonistic actions of o,p'-DDT were partially reversed with 10-fold higher concentrations of 20beta-S, which is consistent with a hormone receptor-mediated mechanism of DDT action. The finding that low concentrations of a wide range of organic environmental contaminants can interfere with a rapid, nongenomic steroid action suggests that this mechanism of endocrine disturbance is of toxicological importance.
Collapse
Affiliation(s)
- Peter Thomas
- The University of Texas at Austin Marine Science Institute, Port Aransas, Texas 78373, USA.
| | | |
Collapse
|
37
|
Mandoki JJ, Mendoza-Patiño N, Molina-Guarneros JA, Jiménez-Orozco FA, Velasco-Velázquez MA, García-Mondragón MJ. Hormone multifunctionalities: a theory of endocrine signaling, command and control. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2004; 86:353-77. [PMID: 15302204 DOI: 10.1016/j.pbiomolbio.2003.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A theory is presented outlining how organisms can function and benefit from multifunctionality of hormones in order to enhance greatly the information-carrying potential of endocrine signaling. Hormones are produced continuously as micropulses, and intermittently as larger pulses. It is generally believed that micropulses generate fluctuating basal hormone concentrations, which may consistently elicit particular responses among diverse variables. Evidence is discussed suggesting that in contrast to the hormone micropulses, the larger endogenous hormone pulses may elicit responses which may differ from one pulse to another and may therefore serve different physiological functions. In this paper we postulate that an endogenous hormone pulse is a specific form of a multisignal message that serves a certain physiological function. Different pulses of a hormone may be signals of diverse multisignal messages that serve different functions. A multisignal message may elicit congruous responses by selectively enhancing some actions and suppressing other actions of the component signals. Various roles of signals of multisignal messages are discussed, as well as processes that may be involved in the diversity and selectivity of actions of different pulses of a hormone. Hormones also are converted into other hormones; we analyze how precursor and derived hormones may function independently of each other, and how precursor hormones may give rise to permissive effects. Mechanisms involved in therapeutic and adverse effects of hormone administrations are analyzed, and a strategy is suggested for developing more selective hormonal therapies.
Collapse
Affiliation(s)
- Juan José Mandoki
- Facultad de Medicina, Departamento de Farmacología, Universidad Nacional Autónoma de México, DF, CP 04510, Apdo. Postal 70-297, Mexico.
| | | | | | | | | | | |
Collapse
|
38
|
Bulayeva NN, Watson CS. Xenoestrogen-induced ERK-1 and ERK-2 activation via multiple membrane-initiated signaling pathways. ENVIRONMENTAL HEALTH PERSPECTIVES 2004; 112:1481-7. [PMID: 15531431 PMCID: PMC1325963 DOI: 10.1289/ehp.7175] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Xenoestrogens can mimic or antagonize the activity of physiological estrogens, and the suggested mechanism of xenoestrogen action involves binding to estrogen receptors (ERs). However, the failure of various in vitro or in vivo assays to show strong genomic activity of xenoestrogens compared with estradiol (E2) makes it difficult to explain their ability to cause abnormalities in animal (and perhaps human) reproductive functions via this pathway of steroid action. E2 has also been shown to initiate rapid intracellular signaling, such as changes in levels of intracellular calcium, cAMP, and nitric oxide, and activations of a variety of kinases, via action at the membrane. In this study, we demonstrate that several xenoestrogens can rapidly activate extracellular-regulated kinases (ERKs) in the pituitary tumor cell line GH3/B6/F10, which expresses high levels of the membrane receptor for ER-alpha (mER). We tested a phytoestrogen (coumestrol), organochlorine pesticides or their metabolites (endosulfan, dieldrin, and DDE), and detergent by-products of plastics manufacturing (p-nonylphenol and bisphenol A). These xenoestrogens (except bisphenolA) produced rapid (3-30 min after application), concentration (10(-14)-10(-8) M)-dependent ERK-1/2 phosphorylation but with distinctly different activation patterns. To identify signaling pathways involved in ERK activation, we used specific inhibitors of ERs, epidermal growth factor receptors, Ca2+ signaling, Src and phosphoinositide-3 kinases, and a membrane structure disruption agent. Multiple inhibitors blocked ERK activation, suggesting simultaneous use of multiple pathways and complex signaling web interactions. However, inhibitors differentially affected each xenoestrogen response examined. These actions may help to explain the distinct abilities of xenoestrogens to disrupt reproductive functions at low concentrations.
Collapse
Affiliation(s)
- Nataliya N Bulayeva
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston, Texas 77555-0645, USA
| | | |
Collapse
|
39
|
Koldzic-Zivanovic N, Seitz PK, Watson CS, Cunningham KA, Thomas ML. Intracellular signaling involved in estrogen regulation of serotonin reuptake. Mol Cell Endocrinol 2004; 226:33-42. [PMID: 15489003 DOI: 10.1016/j.mce.2004.07.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2004] [Revised: 07/06/2004] [Accepted: 07/08/2004] [Indexed: 11/15/2022]
Abstract
17beta-estradiol (E2) regulates neuronal activity via genomic and rapid, non-genomic mechanisms. The rat serotonergic neuronal cell line (RN46A) was used to investigate the rapid effects of E2 on serotonin (5-HT) reuptake and on potential intracellular signaling pathways. RN46A cells express the serotonin transporter (SERT) and estrogen receptor (ER)beta, but not ERalpha. Fifteen minute E2 treatment (10(-9)M) decreased 5-HT uptake. Intracellular cAMP levels were not increased by 15 min E2 treatment; however, E2 caused an increase in intracellular Ca2+ levels, with a maximum response within the first minute. The response was E2 specific, since other steroids (17alpha-estradiol, testosterone, and progesterone) had no effect. The ER antagonist ICI 182,780 blocked the rapid E2 effects on intracellular Ca2+ levels as did the selective ER modulator tamoxifen. In summary, changes in intracellular Ca2+ levels caused by E2 and mediated through ERbeta may be responsible for observed rapid effects of E2 on SERT activity.
Collapse
Affiliation(s)
- Nina Koldzic-Zivanovic
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1031, USA
| | | | | | | | | |
Collapse
|
40
|
Nadal A, Alonso-Magdalena P, Ripoll C, Fuentes E. Disentangling the molecular mechanisms of action of endogenous and environmental estrogens. Pflugers Arch 2004; 449:335-43. [PMID: 15517344 DOI: 10.1007/s00424-004-1343-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Accepted: 08/30/2004] [Indexed: 01/07/2023]
Abstract
The gonadal hormone 17beta-estradiol is involved in numerous cellular processes. In many cases, 17beta-estradiol actions are imitated by synthetic and natural chemicals in the environment. Their actions differ depending on the target tissue, the receptors involved and the molecular pathways activated. The plethora of estrogenic actions is triggered by different receptors and other specific structures that activate different signalling pathways. This amount of information may lead to a maze of effects triggered by endogenous and environmental estrogens that we intend to clarify in this review. Understanding the variety of estrogen receptors, their different locations and the signalling pathways activated by estrogenic ligands is fundamental for understanding the diversity of actions that estrogens have in different tissues and cells.
Collapse
Affiliation(s)
- Angel Nadal
- Institute of Bioengineering, School of Medicine, Miguel Hernández University, Sant Joan Campus, Carretera Alicante-Valencia Km 87, 03550 Alicante, Spain.
| | | | | | | |
Collapse
|
41
|
Grünfeld HT, Bonefeld-Jorgensen EC. Effect of in vitro estrogenic pesticides on human oestrogen receptor α and β mRNA levels. Toxicol Lett 2004; 151:467-80. [PMID: 15261991 DOI: 10.1016/j.toxlet.2004.03.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Revised: 03/15/2004] [Accepted: 03/22/2004] [Indexed: 11/15/2022]
Abstract
Nine widely distributed pesticides were recently demonstrated to possess potential estrogenic properties in oestrogen receptor (ER) transactivation and/or E-screen assays. We tested the effect of these nine pesticides on the human ERalpha and ERbeta mRNA steady state levels in the mamma cancer fibroblast MCF-7BUS cells using on-line RT-PCR. Like 17beta-oestradiol (E2), fenarimol significantly decreased the ERalpha and increased the ERbeta mRNA level. Endosulfan and pirimicarb alone decreased the ERalpha mRNA level weakly. After co-exposure with E2, all the tested pesticides counteracted the E2-induced decrease of the ERalpha mRNA level, but only significantly for prochloraz, dieldrin, and tolchlofos-methyl. Alone no pesticides affected the ERbeta mRNA level significantly, but chlorpyrifos increased the mRNA level weakly. Co-exposure with E2 elicited a significant increased ERbeta mRNA level by prochloraz, fenarimol, endosulfan, dieldrin, and tolchlofos-methyl, whereas no significant effect of the carbamate pesticides on the ERbeta mRNA level was observed. This study demonstrated that organochlor and organophosphorous pesticides possess the ability to interfere with the ERalpha and ERbeta mRNA steady state levels.
Collapse
Affiliation(s)
- H T Grünfeld
- Unit of Environmental Biotechnology, Department of Environmental and Occupational Medicine, University of Aarhus, Vennelyst Boulevard 6, DK-8000 Aarhus, Denmark
| | | |
Collapse
|
42
|
Song RX, Barnes CJ, Zhang Z, Bao Y, Kumar R, Santen RJ. The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor alpha to the plasma membrane. Proc Natl Acad Sci U S A 2004; 101:2076-81. [PMID: 14764897 PMCID: PMC357054 DOI: 10.1073/pnas.0308334100] [Citation(s) in RCA: 238] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Our previous studies demonstrated that 17beta-estradiol (E2) rapidly induces the interaction of estrogen receptor alpha (ERalpha) with the adapter protein Shc, the translocation of ERalpha to the cell membrane, and the formation of dynamic membrane structures in MCF-7 breast cancer cells. The present study examined how E2 causes ERalpha to translocate to the region of the plasma membrane and focused on mechanisms whereby Shc and the insulin-like growth factor-1 receptor (IGF-1R) mediate this process. Shc physically interacts with IGF-1R in the plasma membrane, and E2 activates IGF-1R. We reasoned that ERalpha, when bound to Shc, would be directed to the region of the plasma membrane by the same processes, causing membrane translocation of Shc. We confirmed that E2 rapidly induced IGF-1R phosphorylation and demonstrated that E2 induced formation of a ternary protein complex among Shc, ERalpha, and IGF-1R. Knock down of Shc with a specific small inhibitory RNA decreased the association of ERalpha with IGF-1R by 87%, suggesting that Shc is a crucial molecule in the formation of this ternary complex. Confocal microscopy studies provided further confirmation of the functional roles of Shc and the IGF-1R in the translocation of ERalpha to the region of the membrane. Down-regulation of Shc, ERalpha, or IGF-1R with specific small inhibitory RNAs all blocked E2-induced mitogen-activated protein kinase phosphorylation. Together, our results demonstrate that Shc and IGF-1R serve as key elements in the translocation of ERalpha to the cell membrane and in the facilitation of ERalpha-mediated rapid E2 action.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/metabolism
- Cell Line, Tumor
- Cell Membrane/metabolism
- Estradiol/pharmacology
- Estrogen Receptor alpha
- Humans
- Macromolecular Substances
- Mitogen-Activated Protein Kinases/metabolism
- Phosphorylation/drug effects
- Protein Binding
- Protein Transport
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Shc Signaling Adaptor Proteins
- Src Homology 2 Domain-Containing, Transforming Protein 1
Collapse
Affiliation(s)
- Robert X Song
- Department of Internal Medicine and Biomolecular Research Facility, University of Virginia School of Medicine, Charlottesville, VA 22903, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Nemere I, Pietras RJ, Blackmore PF. Membrane receptors for steroid hormones: signal transduction and physiological significance. J Cell Biochem 2003; 88:438-45. [PMID: 12532320 DOI: 10.1002/jcb.10409] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Membrane receptors for steroid hormones affect signaling pathways that modulate nuclear function, influence neuronal activity, ion flow, and the circulatory system. Indeed, 'new' steroid hormones have been identified by their interaction with membrane-initiated signaling systems. A brief summary of the FASEB Summer Research Conference devoted to these topics is presented in this mini-review. In addition, attendees of the meeting propose introduction of the following terminology: membrane-initiated steroid signaling (MISS) and nuclear-initiated steroid signaling (NISS) to replace more inaccurate terms in current use.
Collapse
Affiliation(s)
- Ilka Nemere
- Department of Nutrition and Food Sciences and the Biotechnology Center, Utah State University, Logan 84322, USA.
| | | | | |
Collapse
|
44
|
Khosravi S, Leung PCK. Differential regulation of gonadotropin-releasing hormone (GnRH)I and GnRHII messenger ribonucleic acid by gonadal steroids in human granulosa luteal cells. J Clin Endocrinol Metab 2003; 88:663-72. [PMID: 12574197 DOI: 10.1210/jc.2002-020866] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
In humans, reproduction was generally believed to be controlled by only one form of GnRH (called mammalian GnRH or GnRHI). However, recently, a second form of GnRH, analogous to chicken GnRHII, was discovered in several tissues, including the human ovary. The regulation and function of GnRHI in the hypothalamus has been well studied. However, the function and regulation of GnRHI, and particularly GnRHII in the ovary, is less well understood. Because gonadal sex steroids are one of the main regulators of reproduction, we investigated, in the present study, the regulation of GnRHI and GnRHII mRNA expression by 17beta-estradiol (E2) and RU486 (a progesterone antagonist) in human granulosa luteal cells (hGLCs). The levels of the mRNA transcripts encoding the two GnRH forms were examined using semiquantitative RT-PCR followed by Southern blot analysis. With time in culture, GnRHI and GnRHII mRNA levels significantly increased, by 120% and 210%, at d 8 and d 1, respectively. The levels remained elevated until the termination of these experiments at d 10. A 24-h treatment of hGLCs with E2 (10(-9) to 10(-7) M) resulted in a dose-dependent decrease and increase in mRNA expression of GnRHI and GnRHII, respectively. E2 (10(-9) M) significantly decreased GnRHI mRNA levels (by 55%) and increased GnRHII mRNA levels (by 294%). Time-course studies demonstrated that E2 (10(-9) M) significantly decreased GnRHI mRNA levels in a time-dependent manner, with maximal inhibition of 77% at 48 h. In contrast, GnRHII mRNA levels significantly increased in a time-dependent fashion, reaching a maximum level of 280% at 24 h. Cotreatment of hGLCs with E2 and tamoxifen (an E2 antagonist) reversed the inhibitory and stimulatory effects of E2 on the mRNA expression of GnRHI and GnRHII, respectively. Time- and dose-dependent treatment with RU486 did not affect GnRHI mRNA levels in hGLCs. In contrast, RU486 treatment significantly increased GnRHII mRNA levels in hGLCs in a time- and dose-dependent fashion, with a maximum increase being observed at 24 h (with 10(-5)M RU486). In summary, the present study demonstrated that the expression of GnRHI and GnRHII at the transcriptional level is differently regulated by E2 and P4 in hGLCs.
Collapse
Affiliation(s)
- Shahram Khosravi
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5
| | | |
Collapse
|
45
|
Kloas W. Amphibians as a model for the study of endocrine disruptors. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 216:1-57. [PMID: 12049206 DOI: 10.1016/s0074-7696(02)16002-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Evidence shows that environmental compounds can interfere with the endocrine systems of wildlife and humans. The main sink of such substances, called endocrine disruptors (EDs), which are mainly of anthropogenic origin, is surface water; thus, aquatic vertebrates such as fishes and amphibians are most endangered. Despite numerous reports on EDs in fishes, information about EDs in amphibians is scarce, and this paucity of information is of particular concern in view of the worldwide decline of amphibians. EDs could contribute to changes of amphibian populations via adverse effects on reproduction and the thyroid system. In amphibians, EDs can affect reproduction by (anti)estrogenic and (anti)androgenic modes of action that produce severe effects including abnormal sexual differentiation. ED actions on the thyroid system cause acceleration or retardation of metamorphosis, which may also affect population levels. Our broad knowledge of amphibian biology and endocrinology indicates that amphibians are very suitable models for the study of EDs. In particular, effects of EDs on the thyroid system triggering metamorphosis can be determined easily and most sensitively in amphibians compared to other vertebrates. A new classification of EDs according to their biological modes of action is proposed because EDs have quite heterogeneous chemical structures, which do not allow prediction of their biological effects. Methods and strategies are proposed for identification and risk assessment of EDs, whether as pure test substances or as mixtures from environmental samples. Effects of EDs on the thyroid system of amphibians can be assessed by a single animal model (Xenopus laevis), whereas the various types of reproduction need comparative studies to investigate whether general endocrine principles do exist among several species of anurans and urodeles. Thus, at least one anuran and one urodelean model are needed to determine ED interference with reproduction.
Collapse
Affiliation(s)
- Werner Kloas
- Department of Inland Fisheries, Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Berlin, Germany
| |
Collapse
|
46
|
CAMPBELL CELESTEH, BULAYEVA NATALIYA, BROWN DAVIDB, GAMETCHU BAHIRU, WATSON CHERYLS. Regulation of the membrane estrogen receptor-alpha: role of cell density, serum, cell passage number, and estradiol. FASEB J 2002; 16:1917-27. [PMID: 12468456 PMCID: PMC1266276 DOI: 10.1096/fj.02-0182com] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We used modified immunocytochemical conditions to quantify a membrane form of estrogen receptor-alpha (mERalpha) in a rat pituitary tumor cell line, GH3/B6/F10. We studied the regulation of mERalpha vs. levels of intracellular ERalpha (iERalpha) using our 96-well plate immunoassay. The anti-ERalpha antibody C542 was used to label the ERalpha (via conjugated alkaline phosphatase) in fixed permeabilized (for iERalpha) vs. nonpermeabilized cells (for mERalpha). Expression of mERalpha was highest at low cell densities (<1000 cells/well) and decreased significantly at densities where cellular processes touched, whereas the more abundant iERalpha increased with increasing cell density over the same range. Serum starvation for 48 h caused increases in mERalpha, whereas iERalpha levels showed no significant changes. A large decline in mERalpha and iERalpha levels with cell passage number was observed. Minutes after nM 17beta-estradiol (E2) treatment, a portion of the cells rounded up and detached from the culture plate, whereas nM cholesterol had no such effect. Although E2 treatment did not change mERalpha levels, the antigen was reorganized from a fine particulate to aggregation into asymmetric large granules of staining. That common culturing conditions favor down-regulation of mERalpha may explain the relatively few reports of this protein in other experimental systems.
Collapse
Affiliation(s)
- CELESTE H. CAMPBELL
- Human Biological Chemistry and Genetics Department, University of Texas, Medical Branch, Galveston Texas, USA; and
| | - NATALIYA BULAYEVA
- Human Biological Chemistry and Genetics Department, University of Texas, Medical Branch, Galveston Texas, USA; and
| | - DAVID B. BROWN
- Human Biological Chemistry and Genetics Department, University of Texas, Medical Branch, Galveston Texas, USA; and
| | - BAHIRU GAMETCHU
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - CHERYL S. WATSON
- Human Biological Chemistry and Genetics Department, University of Texas, Medical Branch, Galveston Texas, USA; and
- Correspondence: Department of Human Biological Chemistry and Genetics, University of Texas, Medical Branch, Galveston, TX 77555-0645, USA. E-mail:
| |
Collapse
|
47
|
Quesada I, Fuentes E, Viso-León MC, Soria B, Ripoll C, Nadal A. Low doses of the endocrine disruptor bisphenol-A and the native hormone 17beta-estradiol rapidly activate transcription factor CREB. FASEB J 2002; 16:1671-3. [PMID: 12207000 DOI: 10.1096/fj.02-0313fje] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endocrine-disrupting chemicals (EDCs) are hormone-like agents present in the environment that alter the endocrine system of wildlife and humans. Most EDCs have potencies far below those of the natural hormone 17beta-E2 when acting through the classic estrogen receptors (ERs). Here, we show that the environmental estrogen Bisphenol-A and the native hormone 17beta-E2 activate the transcription factor, cAMP-responsive element binding protein (CREB) with the same potency. Phosphorylated CREB (P-CREB) was increased after only a 5-minute application of either BPA or 17beta-E2 in a calcium-dependent manner. The effect was reproduced by the membrane-impermeable molecule E2 conjugated to horseradish peroxidase (E-HRP). The increase in P-CREB was not modified by the anti-estrogen ICI 182,780. Therefore, low-dose of BPA activates the transcription factor CREB via an alternative mechanism, involving a non-classical membrane estrogen receptor. Because these effects are elicited at concentrations as low as 10(-9) M, this observation is of environmental and public health relevance.
Collapse
Affiliation(s)
- Ivan Quesada
- Institute of Bioengineering, Miguel Hernández University, Campus of San Juan, Alicante 03550, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Kampa M, Papakonstanti EA, Hatzoglou A, Stathopoulos EN, Stournaras C, Castanas E. The human prostate cancer cell line LNCaP bears functional membrane testosterone receptors that increase PSA secretion and modify actin cytoskeleton. FASEB J 2002; 16:1429-31. [PMID: 12205037 DOI: 10.1096/fj.02-0131fje] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent findings have shown that, in addition to the genomic action of steroids, through intracellular receptors, short-time effects could be mediated through binding to membrane sites. In the present study of prostate cancer LNCaP cells, we report that dihydrotestosterone and the non-internalizable analog testosterone-BSA increase rapidly the release of prostate-specific antigen (PSA) in the culture medium. Membrane testosterone binding sites were identified through ligand binding on membrane preparations, flow cytometry, and confocal laser microscopy of the non-internalizable fluorescent analog testosterone-BSA-FITC, on whole cells. Binding on these sites is time- and concentration-dependent and specific for testosterone, presenting a KD of 10.9 nM and a number of 144 sites/mg protein (approximately 13000 sites/cell). Membrane sites differ immunologically for intracellular androgen receptors. The secretion of PSA after membrane testosterone receptor stimulation was inhibited after pretreatment with the actin cytoskeleton disrupting agent cytochalasin B. In addition, membrane testosterone binding modifies the intracellular dynamic equilibrium of monomeric to filamentous actin and remodels profoundly the actin cytoskeleton organization. These results are discussed in the context of a possible involvement of these sites in cancer chemotherapy.
Collapse
Affiliation(s)
- Maeilena Kampa
- Laboratories of, Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, GR-71110, Greece
| | | | | | | | | | | |
Collapse
|
49
|
Song RX, Santen RJ, Kumar R, Adam L, Jeng MH, Masamura S, Yue W. Adaptive mechanisms induced by long-term estrogen deprivation in breast cancer cells. Mol Cell Endocrinol 2002; 193:29-42. [PMID: 12160999 DOI: 10.1016/s0303-7207(02)00093-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical observations suggest that human breast tumors can adapt in response to endocrine therapy by developing hypersensitivity to estradiol. To understand the mechanisms responsible, we examined estrogenic stimulation of cell proliferation in a model system and provided evidence that long-term deprivation of estradiol causes adaptive hypersensitivity. The enhanced responses to estradiol do not involve mechanisms acting at the level of transcription of estrogen regulated genes. We found no evidence of hypersensitivity when examining the effects of estradiol on regulation of c-myc, pS2, progesterone receptor, several ER reporter genes or c-myb in hypersensitive cells. On the other hand, deprivation of breast cells long term was found to up-regulate a separate pathway whereby the estrogen receptor co-opts a classical growth factor pathway and induces rapid non-genomic effects. Through this pathway, estradiol caused rapid activation of mitogen-activated protein (MAP) kinase. In exploring the mechanisms mediating this event, we found that estradiol binds to cell membrane associated estrogen receptors and causes phosphorylation of Shc, an adaptor protein usually involved in growth factor signaling pathways. ERalpha was found to complex with Shc under these conditions. In turn, Shc bound Grb-2 and Sos which resulted in the activation of MAP kinase. The pure antiestrogen, ICI 182,780, blocked several steps in the rapidly responding ER alpha, Shc, MAP kinase pathway. These non-genomic effects of estradiol produced biologic effects by activating Elk and by inducing morphologic changes in cell membranes. Using confocal microscopy, we demonstrated that estradiol caused a rapid alteration in membrane ruffling, the formation of pseudopodia and translocation of ER alpha to regions contiguous with the cell membrane. These morphologic effects could be blocked with a pure anti-estrogen. We conclude that long-term estradiol deprived cells utilize both genomic (transcriptional) and rapid, non-genomic estradiol induced pathways. We postulate that synergy between these two pathways acting at the level of the cell cycle is responsible for adaptive hypersensitivity.
Collapse
Affiliation(s)
- R X Song
- Department of Medicine, University of Virginia Health Sciences System, Charlottesville, VA, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Many studies have demonstrated the nuclear forms of steroid receptors and their activities, while fewer investigators have identified and described the membrane forms of these receptors. Our immuno-identification approaches for the qualitative and quantitative comparison of the membrane form of the estrogen receptor-alpha (mER alpha) to its nuclear counterpart now allow us to address questions about the comparative levels and regulation of these receptor forms. ER alpha-specific antisense oligonucleotides eliminate mER alpha expression, while only mildly reducing the nuclear ER alpha. Success of immuno-identification for the mER alpha is very sensitive to different fixation protocols, affecting cell permeability (and thus distinction from the intracellular form) and differential epitope preservation. All such identifications must be accompanied by proof of cell membrane integrity and focal plane assessments. The mER alpha expression on selected cells declines rapidly with cell passage number and cell density. Expression of mER alpha is enhanced by serum starvation and selection for specific phases of the cell cycle. The hinge region of the protein is sensitive to ligand-induced epitope masking and to antibody-induced changes in receptor-mediated responses. Responsive cells are often diluted within cell populations by loss of the membrane receptor form. The bimodality of the rapid estrogen action, with inhibitory doses between picomolar and nanomolar stimulatory concentrations, requires detailed dose-response curves. Finally, responsive cells can be lost from assays, as upon estrogen treatment they rapidly round up and leave the substrates to which they are attached. These regulatory phenomena demonstrate that levels of the membrane form of the estrogen receptor are very dynamic.
Collapse
Affiliation(s)
- Cheryl S Watson
- Human Biological Chemistry and Genetics Department, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | | | |
Collapse
|