201
|
Guerrero-Bosagna C, Sabat P, Valladares L. Environmental signaling and evolutionary change: can exposure of pregnant mammals to environmental estrogens lead to epigenetically induced evolutionary changes in embryos? Evol Dev 2005; 7:341-50. [PMID: 15982371 DOI: 10.1111/j.1525-142x.2005.05033.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
DNA methylation is one of the epigenetic and hereditary mechanisms regulating genetic expression in mammalian cells. In this review, we propose how certain natural agents, through their dietary consumption, could induce changes in physiological aspects in mammalian mothers, leading to alterations in DNA methylation patterns of the developing fetus and to the emergence of new phenotypes and evolutionary change. Nevertheless, we hypothesize that this process would require (i) certain key periods in the ontogeny of the organism where the environmental stimuli could produce effects, (ii) particular environmental agents as such stimuli, and (iii) that a genomic persistent change be consequently produced in a population. Depending on the persistence of the environmental stimuli and on whether the affected genes are imprinted genes, induced changes in DNA methylation patterns could become persistent. Moreover, some fragments could be more frequently methylated than others over several generations, leading to biased base change and evolutionary consequences.
Collapse
Affiliation(s)
- Carlos Guerrero-Bosagna
- Laboratorio de Hormonas y Receptores, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile.
| | | | | |
Collapse
|
202
|
Heberden C, Reine F, Grosse B, Henry C, Zagar Y, Chaumaz G, Lieberherr M. Detection of a raft-located estrogen receptor-like protein distinct from ER alpha. Int J Biochem Cell Biol 2005; 38:376-91. [PMID: 16263324 DOI: 10.1016/j.biocel.2005.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 08/30/2005] [Accepted: 09/08/2005] [Indexed: 11/20/2022]
Abstract
17Beta-estradiol (17beta-E2) elicits at the cell membrane rapid actions that remain insensitive to the inhibitory effect of ICI 182,780, a pure estrogen antagonist, and therefore cannot be attributed to the classic nuclear receptors. We addressed the question of the identity of the protein involved in these rapid actions. We first examined the responses of several cell lines for intracellular calcium mobilization, an effect not inhibited by ICI 182,780, tamoxifen and raloxifen. We then demonstrated the presence of binding sites in the membranes, by incubating them with antibodies directed against different domains of ER alpha, and by flow cytometry analysis. The membrane proteins were eluted by affinity chromatography using E2 conjugated to bovine serum albumin as a ligand. Western blots of the elution fractions using an antibody directed against the ligand binding site of ER alpha showed the existence of a protein of approximately 50 kDa. The protein was concentrated in the lipid rafts, together with another heavier form of approximately 66 kDa. The 50 kDa protein was immunoprecipitable, and co-immunoprecipitation experiments showed that it was associated with the Gbeta(1-4) protein, but not with caveolin-1. The protein was expressed in ER alpha-null cells, like HO-23 and Cos-7 cells. Therefore, in the lipid rafts, there exists a protein, similar to, but molecularly distinct from ER alpha.
Collapse
Affiliation(s)
- Christine Heberden
- Laboratoire de Nutrition et Sécurité Alimentaire, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | | | | | | | | | | | | |
Collapse
|
203
|
Viso-León MC, Ripoll C, Nadal A. Oestradiol rapidly inhibits Ca2+ signals in ciliary neurons through classical oestrogen receptors in cytoplasm. Pflugers Arch 2005; 449:33-41. [PMID: 15258764 DOI: 10.1007/s00424-004-1308-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Oestrogen plays a key role in a great variety of actions in the nervous system, either through classical or alternative pathways. The classical pathways are initiated after oestrogen binding to the oestrogen receptors ERalpha or ERbeta, which translocate from the cytoplasm to the nucleus and act there as transcription factors. Alternative pathways are initiated at the plasma membrane and cytoplasm, via binding to classical or non-classical ERs. Using isolated ciliary ganglion neurons from the chick embryo and Ca2+ imaging, we demonstrated that a 10-min exposure to 17beta-oestradiol reduces Ca2+ influx through the plasma membrane. This effect was not reproduced by oestradiol conjugated to bovine serum albumin, which does not cross the plasma membrane, indicating that 17beta-oestradiol was acting intracellularly. ERalpha was detected in the cytoplasm by immunostaining and its involvement in the regulation of Ca2+ influx by ICI182,780 inhibition. The phosphatidylinositol-3 kinase (Pi3-kinase) inhibitor wortmannin and the nitric oxide synthase (NOS) inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME) both blocked the oestradiol effect. The oestradiol effect was reproduced by 8Br-cGMP and abolished in the presence of the cGMP-dependent protein kinase (PKG) inhibitor KT5823. Our study indicates that 17beta-oestradiol can regulate Ca2+ influx via PI3-kinase, NOS and PKG after activation of cytoplasmic ER.
Collapse
Affiliation(s)
- M Carmen Viso-León
- Institut de Bioenginyeria, Universitat Miguel Hernández d'Elx, Campus de Sant Joan, Carretera Alacant-Valéncia Km 87, 03550 Sant Joan d'Alacant, Spain
| | | | | |
Collapse
|
204
|
Mizota K, Yoshida A, Uchida H, Fujita R, Ueda H. Novel type of Gq/11 protein-coupled neurosteroid receptor sensitive to endocrine disrupting chemicals in mast cell line (RBL-2H3). Br J Pharmacol 2005; 145:545-50. [PMID: 15821754 PMCID: PMC1576165 DOI: 10.1038/sj.bjp.0706213] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1 Agonistic neurosteroids, including pregnenolone, dehydroepiandrosterone and its sulfate (DHEAS), caused rapid degranulation in measurements of beta-hexosaminidase (beta-HEX) release from a mast cell line, RBL-2H3. This degranulation was blocked by BSA-conjugated progesterone (PROG-BSA) or 17beta-estradiol, both of which are antagonistic neurosteroids. 2 DHEAS-induced beta-HEX release was blocked by U-73122 or xestospongin C, but not by PTX or EGTA. DHEAS-induced beta-HEX release was also abolished by G(q/11)-AS, but not by G(q/11)-MS. Pharmacological analyses revealed that the neurosteroids stimulated a putative membrane receptor through activation of the novel G(q/11) and phospholipase C. 3 While representative endocrine-disrupting chemicals (EDCs) did not show any degranulation or nocifensive actions by themselves, they blocked the DHEAS-induced degranulation. 4 The binding of a PROG-BSA-fluorescein isothiocyanate conjugate (PROG-BSA-FITC) to cells was inhibited by neurosteroids and EDCs. 5 In the algogenic-induced biting and licking responses test, DHEAS caused agonistic nocifensive actions in a dose-dependent manner between 1 and 10 fmol (i.pl.). DHEAS-induced nocifensive actions were abolished by PROG-BSA or nonylphenol. 6 Taken together, these results suggest that a G(q/11)-coupled neurosteroid receptor may regulate the neuroimmunological activity related to sensory stimulation and that some EDCs have antagonistic actions for this receptor.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- Binding, Competitive/drug effects
- Cattle
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Dehydroepiandrosterone/pharmacology
- Dehydroepiandrosterone Sulfate/pharmacology
- Dose-Response Relationship, Drug
- Estradiol/pharmacology
- Estrenes/pharmacology
- Fluorescein-5-isothiocyanate/chemistry
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Leukemia, Basophilic, Acute/metabolism
- Leukemia, Basophilic, Acute/pathology
- Macrocyclic Compounds
- Male
- Mast Cells/drug effects
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice
- Nociceptors/drug effects
- Oligonucleotides, Antisense/pharmacology
- Oxazoles/pharmacology
- Pain/chemically induced
- Pain/prevention & control
- Pertussis Toxin/pharmacology
- Phenols/pharmacology
- Phosphodiesterase Inhibitors/pharmacology
- Pregnenolone/chemistry
- Pregnenolone/metabolism
- Pregnenolone/pharmacology
- Pyrrolidinones/pharmacology
- Receptors, Steroid/metabolism
- Serum Albumin, Bovine/chemistry
- Type C Phospholipases/antagonists & inhibitors
- Type C Phospholipases/metabolism
- beta-N-Acetylhexosaminidases/metabolism
Collapse
Affiliation(s)
- Kaori Mizota
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Akira Yoshida
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Hitoshi Uchida
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Ryousuke Fujita
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Hiroshi Ueda
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
- Author for correspondence:
| |
Collapse
|
205
|
Alonso-Magdalena P, Laribi O, Ropero AB, Fuentes E, Ripoll C, Soria B, Nadal A. Low doses of bisphenol A and diethylstilbestrol impair Ca2+ signals in pancreatic alpha-cells through a nonclassical membrane estrogen receptor within intact islets of Langerhans. ENVIRONMENTAL HEALTH PERSPECTIVES 2005; 113:969-77. [PMID: 16079065 PMCID: PMC1280335 DOI: 10.1289/ehp.8002] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Glucagon, secreted from pancreatic alpha-cells integrated within the islets of Langerhans, is involved in the regulation of glucose metabolism by enhancing the synthesis and mobilization of glucose in the liver. In addition, it has other extrahepatic effects ranging from lipolysis in adipose tissue to the control of satiety in the central nervous system. In this article, we show that the endocrine disruptors bisphenol A (BPA) and diethylstilbestrol (DES), at a concentration of 10(-9) M, suppressed low-glucose-induced intracellular calcium ion ([Ca2+]i) oscillations in alpha-cells, the signal that triggers glucagon secretion. This action has a rapid onset, and it is reproduced by the impermeable molecule estradiol (E2) conjugated to horseradish peroxidase (E-HRP). Competition studies using E-HRP binding in immunocytochemically identified alpha-cells indicate that 17beta-E2, BPA, and DES share a common membrane-binding site whose pharmacologic profile differs from the classical ER. The effects triggered by BPA, DES, and E2 are blocked by the G alpha i- and G alpha o-protein inhibitor pertussis toxin, by the guanylate cyclase-specific inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, and by the nitric oxide synthase inhibitor N-nitro-L-arginine methyl ester. The effects are reproduced by 8-bromo-guanosine 3',5'-cyclic monophosphate and suppressed in the presence of the cGMP-dependent protein kinase inhibitor KT-5823. The action of E2, BPA, and DES in pancreatic alpha-cells may explain some of the effects elicited by endocrine disruptors in the metabolism of glucose and lipid.
Collapse
Affiliation(s)
- Paloma Alonso-Magdalena
- Institute of Bioengineering, Miguel Hernández University, Sant Joan d'Alacant, Alicante, Spain
| | | | | | | | | | | | | |
Collapse
|
206
|
Arreguin-Arevalo JA, Nett TM. A Nongenomic Action of 17β-Estradiol as the Mechanism Underlying the Acute Suppression of Secretion of Luteinizing Hormone1. Biol Reprod 2005; 73:115-22. [PMID: 15772257 DOI: 10.1095/biolreprod.105.040329] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The objective of the present study was to determine the ability of 17beta-estradiol (E(2)) and conjugated forms of E(2) (E(2) conjugated to BSA [E(2)-BSA] and a novel conjugate, E(2) conjugated to a small peptide [E(2)-PEP]) to prevent the GnRH-induced secretion of LH and to determine the role of estradiol receptors (ERs) and ER subtypes (ERalpha, also known as ESR1, and ERbeta, also known as ESR2) in the mediation of the acute action of E(2) in primary cultures of ovine pituitary cells. Preincubation of cells for 15 min with E(2), E(2)-BSA, or E(2)-PEP prevented the GnRH-induced secretion of LH (P < 0.01). Treatment of cells with nonestrogenic steroid hormones did not affect secretion of LH when given alone, nor did these steroids impair the E(2)-induced inhibition of LH secretion (P > 0.1). Likewise, treatment of cells with the ER-antagonists tamoxifen, hydroxytamoxifen, or ICI 182 780 did not affect (P > 0.1) secretion of LH when given alone but did prevent (P < 0.01) the inhibition by E(2) and the E(2)-conjugates on GnRH-induced secretion of LH. When cells were treated with subtype-selective ER agonists, the ERalpha agonist (propylpyrazole-triol), but not the ERbeta agonist (diarylpropionitrile), decreased (P < 0.01) the GnRH-induced secretion of LH. In conclusion, the rapidity by which E(2) prevented GnRH-induced release of LH in ovine pituitary cells suggests that this inhibition is mediated via a nongenomic action of E(2). The inhibition of GnRH-induced secretion of LH proved to be steroid specific and mediated by ERs. It may occur specifically through ERalpha. The fact that E(2)-BSA or E(2)-PEP mimicked the action of E(2) suggests that this effect was mediated by an ER associated with the plasma membrane.
Collapse
|
207
|
Ballinger SW. Mitochondrial dysfunction in cardiovascular disease. Free Radic Biol Med 2005; 38:1278-95. [PMID: 15855047 DOI: 10.1016/j.freeradbiomed.2005.02.014] [Citation(s) in RCA: 271] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 02/15/2005] [Accepted: 02/15/2005] [Indexed: 12/11/2022]
Abstract
Whereas the pathogenesis of atherosclerosis has been intensively studied and described, the underlying events that initiate cardiovascular disease are not yet fully understood. A substantial number of studies suggest that altered levels of oxidative and nitrosoxidative stress within the cardiovascular environment are essential in the development of cardiovascular disease; however, the impact of such changes on the subcellular or organellar components and their functions that are relevant to cardiovascular disease inception are less understood. In this regard, studies are beginning to show that mitochondria not only appear susceptible to damage mediated by increased oxidative and nitrosoxidative stress, but also play significant roles in the regulation of cardiovascular cell function. In addition, accumulating evidence suggests that a common theme among cardiovascular disease development and cardiovascular disease risk factors is increased mitochondrial damage and dysfunction. This review discusses aspects relating mitochondrial damage and function to cardiovascular disease risk factors and disease development.
Collapse
Affiliation(s)
- Scott W Ballinger
- Division of Molecular and Cellular Pathology, VH G019F, 1530 3rd Avenue South, Birmingham, AL 35294-0019, USA.
| |
Collapse
|
208
|
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
|
209
|
Vidaeff AC, Sever LE. In utero exposure to environmental estrogens and male reproductive health: a systematic review of biological and epidemiologic evidence. Reprod Toxicol 2005; 20:5-20. [PMID: 15808781 DOI: 10.1016/j.reprotox.2004.12.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Revised: 11/12/2004] [Accepted: 12/04/2004] [Indexed: 10/25/2022]
Abstract
In recent years, chemicals with hormone-like properties have become a topic of scientific and public discussion. It has been hypothesized that prenatal exposure of the male fetus to endocrine disruptors may be responsible for a series of outcomes, such as hypospadias and cryptorchidism. The purpose of this study was to review the endocrine disruption hypothesis, to present the relevant supporting evidence, to summarize the current knowledge, to identify gaps and limitations in the interpretation of published data, and to define future directions in research. An update on environmental estrogens was followed by an assessment of the biological plausibility and evidence connecting the environmental chemicalization with adverse reproductive outcomes in males. Subsequently, we carried out a systematic review of human studies attempting to document a direct effect of exogenous estrogens on the male reproductive system. The results do not support with certainty the view that environmental estrogens contribute to an increase in male reproductive disorders, neither do they provide sufficient grounds to reject such a hypothesis.
Collapse
Affiliation(s)
- Alex C Vidaeff
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, 6431 Fannin, Suite 3.604, Houston, TX 77030, USA.
| | | |
Collapse
|
210
|
Al-Majed HT, Squires PE, Persaud SJ, Huang GCC, Amiel S, Whitehouse BJ, Jones PM. Effect of 17beta-estradiol on insulin secretion and cytosolic calcium in Min6 mouse insulinoma cells and human islets of Langerhans. Pancreas 2005; 30:307-13. [PMID: 15841038 DOI: 10.1097/01.mpa.0000161886.17492.22] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Female gonadal steroids can exert an insulinotropic effect in vivo. The objective of this study was to investigate the effects in vitro of 17-beta-estradiol (17beta-E2) on changes in cytosolic calcium ([Ca]i) and on insulin secretion from the MIN6 mouse insulinoma cell line and human primary islets of Langerhans. METHODS Stimulus-induced changes in [Ca]i were measured in Fura-2-loaded cells by single cell microfluorimetry. The effects of 17beta-E2 on insulin secretion were measured in static incubation experiments, and the rate and pattern of secretory responses were studied in multi-channel perifusion experiments. RESULTS 17Beta-E2 (1-100 nmol/L) enhanced basal (2 mmol/L glucose) insulin secretion but had no effect on secretory responses to 20 mmol/L glucose or to depolarizing stimuli (100 micromol/L tolbutamide, 20 mmol/L KCl). Approximately 60% of MIN6 cells responded to 17beta-E2 (1-100 nmol/L) with a small but sustained increase in [Ca]i, whereas 98% of MIN6 cells responded to tolbutamide (100 micromol/L). Similar effects were observed in experiments using human primary beta cells. In contrast, 17beta-E2 had no detectable effect on the increases in [Ca]i evoked by tolbutamide (100 micromol/L) or glucose (20 mmol/L). CONCLUSIONS Our observations are consistent with a rapid effect of 17beta-E2 to depolarize beta cells leading to an influx of extracellular Ca and the initiation of insulin secretion by the consequent elevations in [Ca]i. We suggest that this may offer a mechanism through which circulating estradiol can influence beta-cell responsiveness to other signals.
Collapse
Affiliation(s)
- Hana T Al-Majed
- Division of Reproductive Health, Endocrinology and Development, King's College London, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
211
|
Adachi T, Yasuda K, Mori C, Yoshinaga M, Aoki N, Tsujimoto G, Tsuda K. Promoting insulin secretion in pancreatic islets by means of bisphenol A and nonylphenol via intracellular estrogen receptors. Food Chem Toxicol 2005; 43:713-9. [PMID: 15778011 DOI: 10.1016/j.fct.2005.01.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Revised: 01/07/2005] [Accepted: 01/19/2005] [Indexed: 11/29/2022]
Abstract
In this study, we investigated the effects of endocrine disrupters bisphenol A (BPA) and nonylphenol (NP) on insulin secretion from rat pancreatic islets. Following acute exposure to BPA and NP, neither BPA nor NP (0.1, 1, 10, 100 and 1000 microg/l) affected insulin secretion in concentrations of 16.7 mM glucose. However, insulin secretion following long-term exposure to BPA or NP for 24 h in 16.7 mM glucose was significantly higher than without exposure. To determine whether increased insulin secretion resulting from long-term exposure to BPA and NP is induced via intracellular estrogen receptors, we blocked the cytosolic/nuclear estrogen receptors, using actinomycin-D (Act-D), an inhibitor of RNA synthesis, and ICI 182,780 (ICI), an estrogen receptor inhibitor. Following long-term exposure to BPA (10 microg/l) or NP (10 microg/l), Act-D or ICI treatment eliminated the facilitation of insulin secretion. In conclusion, we have demonstrated for the first time that long-term exposure to endocrine disrupters, such as BPA and NP, promotes in vitro insulin secretion from the pancreatic islets, via cytosolic/nuclear estrogen receptors.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
212
|
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
|
213
|
Cassidy RA, Natarajan S, Vaughan GM. The link between the insecticide heptachlor epoxide, estradiol, and breast cancer. Breast Cancer Res Treat 2005; 90:55-64. [PMID: 15770527 DOI: 10.1007/s10549-004-2755-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Given the suspected effects of estrogens on breast cancer, xenoestrogenic insecticides may be a risk factor. Studies of the weak xenoestrogen, 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE), have failed to demonstrate a causal relationship, though another estrogenic organochlorine insecticide, dieldrin, belonging to the cyclodiene family, has recently been linked to breast cancer. Other cyclodienes such as heptachlor epoxide (HE) and oxychlordane (OC) present in breast tissue have not been evaluated as rigorously, presumably due to their lower concentration and lower recovery using solvent extraction procedures. We used sparging extraction coupled with gas chromatography to determine the levels of HE, OC, and DDE in adipose tissue within breast biopsies in a series of 34 women evaluated for breast abnormality. Of the three insecticides tested, only HE (p=0.007) was positively associated with prevalence of breast cancer in the biopsies. In rapid, non-genomic studies using isolated human leukocytes, flow cytometric methods were used to measure HE-induced oxidants and DNA damage. These studies indicated that HE, at concentrations similar to those in breast biopsies, induced an inverted-U increase in intracellular oxidants and DNA strand breaks [both blocked by specific nitric oxide- (NO-) synthesis blockade withL: -NMMA] in human polymorphonuclear leukocytes (PMNs). HE-treated PMNs also induced damage to surrounding lymphocytes in mixed-leukocyte incubations (also inhibited by NO blockade). The HE-induced changes in NO were inhibited by 17beta-estradiol-(17beta-E2) receptor antagonists and were mimicked by similar concentrations of 17beta-E2. The addition of tumor necrosis factor-alpha (TNF-alpha) increased intracellular oxidants and DNA damage and shifted the responses to lower HE concentrations. This study, along with others, suggests that HE-induced NO production may contribute to initiation, promotion, and progression of cancer.
Collapse
|
214
|
Somjen D, Kohen F, Lieberherr M, Gayer B, Schejter E, Katzburg S, Limor R, Sharon O, Knoll E, Posner GH, Kaye AM, Stern N. Membranal effects of phytoestrogens and carboxy derivatives of phytoestrogens on human vascular and bone cells: new insights based on studies with carboxy-biochanin A. J Steroid Biochem Mol Biol 2005; 93:293-303. [PMID: 15860273 DOI: 10.1016/j.jsbmb.2004.12.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Estradiol-17beta (E2) and some phytoestrogens induce a biphasic effect on DNA synthesis in cultured human vascular smooth muscle cells (VSMC), i.e., stimulation at low concentrations and inhibition at high concentrations. These compounds also increase the specific activity of creatine kinase (CK) as well as intracellular Ca2+ concentration in both VSMC and human female-derived cultured bone cells (OBs), and stimulate ERK1/2 phosphorylation in VSMC. At least some of these effects are exerted via membranal binding sites (mER), as would appear from observations that protein-bound, membrane impermeant estrogenic complexes can mimic the effect of E2 on DNA synthesis, intracellular Ca2+ concentration and MAPK, but not on CK activity. We now extend these studies by examining the effects of a novel carboxy-derivative of biochanin A, 6-carboxy-biochanin A (cBA) in VSMC and human osteoblasts in culture. cBA increased DNA synthesis in VSMC in a dose-dependent manner and was able to maintain this effect when linked to a cell membrane impermeable protein. In VSMC both cBA and estradiol, in their free or protein-bound forms induced a steep and immediate rise in intracellular calcium. Both the free and protein-bound conjugates of cBA and estradiol increased net MAPK-kinase activity. Neither the stimulatory effect of cBA nor the inhibitory effect of estradiol on DNA synthesis in VSMC could be shown in the presence of the MAPK-kinase inhibitor UO126. The presence of membrane binding sites for both estradiol and cBA was supported by direct visualization, using fluorescence labeling of their respective protein conjugates, E2-BSA and cBA-ovalbumin. Furthermore, these presumed membrane ER for estradiol and cBA were co-localized. In cultured human osteoblasts, cBA stimulated CK activity in a dose related fashion, which paralleled the increase in CK induced by estradiol per se, confirming the estrogenic properties of cBA in human bone cells. Both the free and protein-bound forms of cBA elicited immediate and substantial increments in intracellular Ca2+, similar to, but usually larger than the responses elicited by estradiol per se. cBA also increased ERalpha and suppressed ERbeta mRNA expression in human osteoblasts. Cultured human osteoblasts also harbor membrane binding sites for protein-bound form of cG, which are co-localized with the binding sites for protein-bound estradiol. The extent to which these properties of the novel synthetic phytoestrogen derivatives may be utilized to avert human vascular and/or bone disease requires further study.
Collapse
Affiliation(s)
- D Somjen
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Centre and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Thomas P, Pang Y, Filardo EJ, Dong J. Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells. Endocrinology 2005; 146:624-32. [PMID: 15539556 DOI: 10.1210/en.2004-1064] [Citation(s) in RCA: 1057] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Although nonclassical estrogen actions initiated at the cell surface have been described in many tissues, the identities of the membrane estrogen receptors (mERs) mediating these actions remain unclear. Here we show that GPR30, an orphan receptor unrelated to nuclear estrogen receptors, has all the binding and signaling characteristics of a mER. A high-affinity (dissociation constant 2.7 nm), limited capacity, displaceable, single binding site specific for estrogens was detected in plasma membranes of SKBR3 breast cancer cells that express GPR30 but lack nuclear estrogen receptors. Progesterone-induced increases and small interfering RNA-induced decreases in GPR30 expression in SKBR3 cells were accompanied by parallel changes in specific estradiol-17beta (E2) binding. Plasma membranes of human embryonic kidney 293 cells transfected with GPR30, but not those of untransfected cells, and human placental tissues that express GPR30 also displayed high-affinity, specific estrogen binding typical of mERs. E2 treatment of transfected cell membranes caused activation of a stimulatory G protein that is directly coupled to the receptor, indicating GPR30 is a G protein-coupled receptor (GPCR), and also increased adenylyl cyclase activity. The finding that the antiestrogens tamoxifen and ICI 182,780, and an environmental estrogen, ortho,para-dichlorodiphenyldichloroethylene (o,p'-DDE), have high binding affinities to the receptor and mimic the actions of E2 has important implications for both the development and treatment of estrogen-dependent breast cancer. GPR30 is structurally unrelated to the recently discovered family of GPCR-like membrane progestin receptors. The identification of a second distinct class of GPCR-like steroid membrane receptors suggests a widespread role for GPCRs in nonclassical steroid hormone actions.
Collapse
Affiliation(s)
- P Thomas
- University of Texas Marine Science Institute, 750 Channel View Drive, Port Aransas, Texas 78373, USA.
| | | | | | | |
Collapse
|
216
|
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
|
217
|
Canesi L, Betti M, Lorusso LC, Ciacci C, Gallo G. 'In vivo' effects of Bisphenol A in Mytilus hemocytes: modulation of kinase-mediated signalling pathways. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2005; 71:73-84. [PMID: 15642633 DOI: 10.1016/j.aquatox.2004.10.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Revised: 10/13/2004] [Accepted: 10/14/2004] [Indexed: 05/24/2023]
Abstract
Endocrine disrupting chemicals (EDCs) include a variety of natural and synthetic estrogens, as well as estrogen-mimicking chemicals. We have previously shown that in the hemocytes of the mussel Mytilus galloprovincialis Lam. both natural and environmental estrogens in vitro can rapidly affect the phosphorylation state of components of tyrosine kinase-mediated cell signalling, in particular of mitogen activated protein kinases (MAPKs) and signal transducers and activators of transcription (STAT), that are involved in mediating the hemocyte immune response. These effects were consistent with the hypothesis that 'alternative' modes of estrogen action involving kinase-mediated pathways similar to those described in mammalian systems are also present in invertebrate cells. This possibility was investigated in vivo with Bisphenol A (BPA): mussels were injected with BPA and hemocytes sampled at 6, 12, and 24 h post-injection. The results show that BPA (25 nM nominal concentration in the hemolymph) lead to a significant lysosomal membrane destabilisation at all times post-injection, indicating BPA-induced stress conditions in the hemocytes, whereas lower concentrations were ineffective. BPA induced significant changes in the phosphorylation state of MAPK and STAT members, as evaluated by SDS-PAGE and WB of hemocyte protein extracts with specific antibodies, although to a different degree at different exposure times. In particular, BPA induced a dramatic decrease in phosphorylation of the stress-activated p38 MAPK, whose activation is crucial in mediating the bactericidal activity. Moreover, BPA decreased the phosphorylation of a CREB-like transcription factor (cAMP-responsive element binding protein). The results demonstrate that BPA can affect kinase-mediated cell signalling in mussel hemocytes also in vivo, and suggest that EDCs may affect gene expression in mussel cells through modulation of the activity of transcription factors secondary to cytosolic kinase cascades. Overall, these data address the importance of investigating full range responses to EDCs in ecologically relevant marine invertebrate species.
Collapse
Affiliation(s)
- Laura Canesi
- Istituto di Scienze Fisiologiche, Università di Urbino Carlo Bo, Loc. Crocicchia, 61029 Urbino PU, Italy.
| | | | | | | | | |
Collapse
|
218
|
Liu D, Homan LL, Dillon JS. Genistein acutely stimulates nitric oxide synthesis in vascular endothelial cells by a cyclic adenosine 5'-monophosphate-dependent mechanism. Endocrinology 2004; 145:5532-9. [PMID: 15319357 DOI: 10.1210/en.2004-0102] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Genistein may improve vascular function, but the mechanism of this effect is unclear. We tested the hypothesis that genistein directly regulates vascular function through stimulation of endothelial nitric oxide synthesis. Genistein activated endothelial nitric oxide synthase (eNOS) in intact bovine aortic endothelial cells and human umbilical vein endothelial cells over an incubation period of 10 min. The maximal eNOS activity was at 1 microm genistein. Consistent with this activation pattern, 1 microm genistein maximally stimulated the phosphorylation of eNOS at serine 1179 at 10 min of incubation. The rapid activation of eNOS by genistein was not dependent on RNA transcription or new protein synthesis and was not blocked by a specific estrogen receptor antagonist. In addition, inhibition of MAPK or phosphatidylinositol 3-OH kinase/Akt kinase had no affect on eNOS activation by genistein. Furthermore, the genistein effect on eNOS was also independent of tyrosine kinase inhibition. However, inhibition of cAMP-dependent kinase [protein kinase A (PKA)] by H89 completely abolished the genistein-stimulated eNOS activation and phosphorylation, suggesting that genistein acted through a PKA-dependent pathway. These findings demonstrated that genistein had direct nongenomic effects on eNOS activity in vascular endothelial cells, leading to eNOS activation and nitric oxide synthesis. These effects were mediated by PKA and were unrelated to an estrogenic effect. This cellular mechanism may underlie some of the cardiovascular protective effects proposed for soy phytoestrogens.
Collapse
Affiliation(s)
- Dongmin Liu
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa and Veterans Affairs Medical Center, Iowa City, Iowa 52246, USA
| | | | | |
Collapse
|
219
|
Tang S, Tan SL, Ramadoss SK, Kumar AP, Tang MHE, Bajic VB. Computational method for discovery of estrogen responsive genes. Nucleic Acids Res 2004; 32:6212-7. [PMID: 15576347 PMCID: PMC535661 DOI: 10.1093/nar/gkh943] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Estrogen has a profound impact on human physiology and affects numerous genes. The classical estrogen reaction is mediated by its receptors (ERs), which bind to the estrogen response elements (EREs) in target gene's promoter region. Due to tedious and expensive experiments, a limited number of human genes are functionally well characterized. It is still unclear how many and which human genes respond to estrogen treatment. We propose a simple, economic, yet effective computational method to predict a subclass of estrogen responsive genes. Our method relies on the similarity of ERE frames across different promoters in the human genome. Matching ERE frames of a test set of 60 known estrogen responsive genes to the collection of over 18,000 human promoters, we obtained 604 candidate genes. Evaluating our result by comparison with the published microarray data and literature, we found that more than half (53.6%, 324/604) of predicted candidate genes are responsive to estrogen. We believe this method can significantly reduce the number of testing potential estrogen target genes and provide functional clues for annotating part of genes that lack functional information.
Collapse
Affiliation(s)
- Suisheng Tang
- Knowledge Extraction Lab, Institute for Infocomm Research, 21 Heng Mui Keng Terrace, Singapore 119613
| | | | | | | | | | | |
Collapse
|
220
|
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
|
221
|
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
|
222
|
Toran-Allerand CD. Estrogen and the brain: beyond ER-α and ER-β. Exp Gerontol 2004; 39:1579-86. [PMID: 15582272 DOI: 10.1016/j.exger.2004.05.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2004] [Accepted: 05/17/2004] [Indexed: 11/25/2022]
Abstract
17beta-Estradiol is a greatly under-appreciated neural growth and trophic factor for the mammalian brain of all ages. Like other growth factors, such as the neurotrophins, 17beta-estradiol influences neurogenesis, neuronal differentiation, and neuronal survival of its targets throughout life. Estrogen elicits developmentally regulated differentiative effects, which are not normally seen in the adult brain. However, re-expression of this developmental response occurs in the adult, following loss of trophic support, whether induced by estrogen deprivation or brain injury. In addition to the classical intranuclear estrogen receptors (ER) ER-alpha and ER-beta, we have recently identified a novel, plasma membrane-associated, putative ER that is neither ER-alpha nor ER-beta, which we have designated 'ER-X'. ER-X is a developmentally regulated estrogen-binding protein, present in wild-type, ER-alpha gene-disrupted (alphaERKO) and ER-alpha null mice, which is re-expressed following ischemic brain injury. The preferred ligand of ER-X is 17alpha-estradiol. Although ER-X shares some homology with the C-terminus of ER-alpha, it is not an alternative splicing variant and may be a new gene. While ER-X appears to mediate 17alpha- and 17beta-estradiol activation of the MAPK cascade, ER-alpha, in contrast, is inhibitory to its activation. Estradiol activation of MAPK/ERK may be particularly relevant for neuroprotection during aging and Alzheimer's disease.
Collapse
Affiliation(s)
- C D Toran-Allerand
- Department of Anatomy and Cell Biology, and Neurology, Centers for Neurobiology and Behavior and Reproductive Sciences, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
223
|
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
|
224
|
Govind AP, Thampan RV. Membrane associated estrogen receptors and related proteins: localization at the plasma membrane and the endoplasmic reticulum. Mol Cell Biochem 2004; 253:233-40. [PMID: 14619974 DOI: 10.1023/a:1026068017309] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The female sex steroid, estradiol 17beta, mediates its effect through its association with estrogen receptor present in the target cell. So far the major emphasis has been given to the genomic actions of the hormone mediated by the nuclear estrogen receptors. Recent years have seen a shift in the ideas revealing the existence of estradiol binding entities both in the plasma membrane and the endoplasmic reticulum. Though the true identity of this membrane associated receptors is far from being known, a functional role for the same have been implicated both at the genomic as well as the non-genomic level. The major focus of the review is to highlight the existence of membrane associated estrogen receptors and receptor-related proteins and the functional roles played by some of them. The signalling events exerted by this class of membrane associated estrogen receptor could partly explain the physiological significance of estrogen in cardiovascular disease, osteoporosis and breast cancer as well as the molecular mechanism associated with xenoestrogen action.
Collapse
Affiliation(s)
- Anitha P Govind
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | |
Collapse
|
225
|
Kanda N, Watanabe S. 17beta-estradiol enhances the production of granulocyte-macrophage colony-stimulating factor in human keratinocytes. J Invest Dermatol 2004; 123:329-37. [PMID: 15245433 DOI: 10.1111/j.0022-202x.2004.23231.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is effective for impaired wound repair. Estrogen is known to enhance wound repair. We examined if 17beta-estradiol (E2) may in vitro enhance GM-CSF production in human keratinocytes. E2 and membrane-impermeable bovine serum albumin-conjugated E2 increased GM-CSF secretion, mRNA stability, and promoter activity. The element homologous to activator protein-1 (AP-1) on the promoter was responsible for the activation. E2 enhanced transcriptional activity and DNA binding of AP-1. E2 transiently generated c-Fos protein, and shifted AP-1 composition from c-Jun homodimers to c-Fos/c-Jun heterodimers in keratinocytes. E2-induced enhancement of GM-CSF secretion, mRNA stability, and promoter activity were not suppressed by estrogen receptor antagonist ICI 182,780, however, suppressed by conventional protein kinase C inhibitor Gö6976 and PD98059, an inhibitor of mitogen-activated protein kinase kinase (MEK). Gö6976 and PD98059 suppressed E2-induced c-Fos expression and enhancement of DNA-binding and transcriptional activity at AP-1. E2 induced membrane translocation of protein kinase Calpha, which was suppressed by phosphatidylinositol (PI)-specific phospholipase C (PLC) inhibitor U73122. E2 stimulated the phosphorylation of extracellular signal-regulated kinase (ERK), which was suppressed by PD98059, Gö6976, and U73122. E2 transiently generated inositol 1,4,5-triphosphate in keratinocytes, which was suppressed by U73122 and guanine nucleotide-binding protein inhibitor. These results suggest that E2 may enhance GM-CSF production via guanine nucleotide-binding protein-coupled membrane receptors and signaling cascade of PI-specific PLC/protein kinase Calpha/MEK/ERK.
Collapse
Affiliation(s)
- Naoko Kanda
- Department of Dermatology, Teikyo University, School of Medicine, Kaga, Itabashi-Ku, Tokyo, Japan.
| | | |
Collapse
|
226
|
Taguchi Y, Koslowski M, Bodenner DL. Binding of estrogen receptor with estrogen conjugated to bovine serum albumin (BSA). NUCLEAR RECEPTOR 2004; 2:5. [PMID: 15318942 PMCID: PMC516042 DOI: 10.1186/1478-1336-2-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2003] [Accepted: 08/19/2004] [Indexed: 11/10/2022]
Abstract
BACKGROUND: The classic model of estrogen action requires that the estrogen receptor (ER) activates gene expression by binding directly or indirectly to DNA. Recent studies, however, strongly suggest that ER can act through nongenomic signal transduction pathways and may be mediated by a membrane bound form of the ER. Estradiol covalently linked to membrane impermeable BSA (E2-BSA) has been widely used as an agent to study these novel membrane-associated ER events. However, a recent report suggests that E2-BSA does not compete for E2 binding to purified ER in vitro. To resolve this apparent discrepancy, we performed competition studies examining the binding of E2 and E2-BSA to both purified ER preparations and ER within intact cells. To eliminate potential artifacts due to contamination of commercially available E2-BSA preparations with unconjugated E2 (usually between 3-5%), the latter was carefully removed by ultrafiltration. RESULTS: As previously reported, a 10-to 1000-fold molar excess of E2-BSA was unable to compete with 3H-E2 binding to ER when added simultaneously. However, when ER was pre-incubated with the same concentrations of E2-BSA, the binding of 3H-E2 was significantly reduced. E2-BSA binding to a putative membrane-associated ER was directly visualized using fluorescein labeled E2-BSA (E2-BSA-FITC). Staining was restricted to the cell membrane when E2-BSA-FITC was incubated with stable transfectants of the murine ERalpha within ER-negative HeLa cells and with MC7 cells that endogenously produce ERalpha. This staining appeared highly specific since it was competed by pre-incubation with E2 in a dose dependent manner and with the competitor ICI-182,780. CONCLUSIONS: These results demonstrate that E2-BSA does bind to purified ER in vitro and to ER in intact cells. It seems likely that the size and structure of E2-BSA requires more energy for it to bind to the ER and consequently binds more slowly than E2. More importantly, these findings demonstrate that in intact cells that express ER, E2-BSA binding is localized to the cell membrane, strongly suggesting a membrane bound form of the ER.
Collapse
Affiliation(s)
- Yasuto Taguchi
- Obstetrics and Gynecology, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Mirek Koslowski
- Endocrinology and Metabolism, Emory University School of Medicine and Veterans Affairs Medical Center, Atlanta, Georgia 30033, USA
| | - Donald L Bodenner
- Geriatrics, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
227
|
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
|
228
|
Abrahám IM, Todman MG, Korach KS, Herbison AE. Critical in vivo roles for classical estrogen receptors in rapid estrogen actions on intracellular signaling in mouse brain. Endocrinology 2004; 145:3055-61. [PMID: 14976146 DOI: 10.1210/en.2003-1676] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Estrogen exerts classical genomic as well as rapid nongenomic actions on neurons. The mechanisms involved in rapid estrogen signaling are poorly defined, and the roles of the classical estrogen receptors (ERs alpha and beta) are unclear. We examined here the in vivo role of classical ERs in rapid estrogen actions by evaluating the estrogen-induced effects on two major signaling pathways within the brains of alphaER-, betaER-, and double alphabetaER-knockout (ERKO) ovariectomized female mice. Estrogen significantly (P < 0.05) increased the numbers of phospho-cAMP response element binding protein (phospho-CREB)-immunoreactive cells in specific brain regions of wild-type mice in a time-dependent manner beginning within 15 min. In brain areas that express predominantly ERbeta, this response was absent in betaERKO mice, whereas brain regions that express mostly ERalpha displayed no change in alphaERKO mice. In the medial preoptic nucleus (MPN), an area that expresses both ERs, the estrogen-induced phosphorylation of CREB was normal in both alphaERKO and betaERKO mice. However, estrogen had no effect on CREB phosphorylation in the MPN, or any other brain region, in double alphabetaERKO animals. Estrogen was also found to increase MAPK phosphorylation levels in a rapid (<15 min) manner within the MPN. In contrast to CREB signaling, this effect was lost in either alphaERKO or betaERKO mice. These data show that ERalpha and ERbeta play region- and pathway-specific roles in rapid estrogen actions throughout the brain. They further indicate an indispensable role for classical ERs in rapid estrogen actions in vivo and highlight the importance of ERs in coordinating both classical and rapid actions of estrogen.
Collapse
Affiliation(s)
- István M Abrahám
- Laboratory of Neuroendocrinology, Babraham Institute, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
229
|
Hamilton KL, Mbai FN, Gupta S, Knowlton AA. Estrogen, heat shock proteins, and NFkappaB in human vascular endothelium. Arterioscler Thromb Vasc Biol 2004; 24:1628-33. [PMID: 15231513 DOI: 10.1161/01.atv.0000137188.76195.fb] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We hypothesized that estrogen would increase HSP72 in human coronary artery endothelial cells (HCAEC), and that these would be more sensitive to estrogen than our previous observations in myocytes. METHODS AND RESULTS HCAEC were treated with 17beta-estradiol or tamoxifen, ranging from physiological to pharmacological(1 nM to 10 micromol/L) for either 24 hours (early) or 7 days (chronic). HSP expression was assessed by Western blots. Both early and chronic 17beta-estradiol and tamoxifen increased HSP72. Electromobility shift assays (EMSA) showed activation of HSF-1 with early, but not chronic, 17beta-estradiol. 17beta-Estradiol activated NFkappaB within 10 minutes, and the ER-alpha selective inhibitor, ICI 182 780, abolished this effect. Transcription factor decoys containing the heat shock element blocked HSP72 induction. Estrogen pretreatment decreased lactate dehydrogenase release with hypoxia. This protective effect persisted despite blockade of HSF-1 by decoys. However, an NF-kappaB decoy prevented the increase in HSP72 and abolished the estrogen-associated protection during hypoxia. CONCLUSIONS 17beta-Estradiol upregulates HSP72 early and chronically via different mechanisms in HCAEC, and provides cytoprotection during hypoxia, independent of HSP72 induction. NF-kappaB mediates the early increase in HSP72, suggesting that estrogen activates NF-kappaB via a nongenomic, receptor-dependent mechanism, and this leads to activation of HSF-1. Activation of NF-kappaB was critical for estrogen-associated protection. Further studies are needed to elucidate the involved signaling pathways. We hypothesized that estrogen would increase HSP72 in human coronary artery endothelial cells (HCAEC). Both early and chronic treatment increased HSP72. EMSA showed activation of HSF-1 with early, but not chronic, 17beta-estradiol. Transcription factor decoys blocked estrogen-related HSP72 induction. Estrogen decreased LDH release with hypoxia. An NF-kappaB decoy blocked the HSP72 increase and estrogen-associated protection.
Collapse
|
230
|
Lee MH, Kim E, Kim TS. Exposure to 4-tert-octylphenol, an environmentally persistent alkylphenol, enhances interleukin-4 production in T cells via NF-AT activation. Toxicol Appl Pharmacol 2004; 197:19-28. [PMID: 15126071 DOI: 10.1016/j.taap.2004.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2003] [Accepted: 02/10/2004] [Indexed: 11/16/2022]
Abstract
4-tert-Octylphenol (OP) is a representative endocrine disruptor that may have adverse effects on human health. The influence of this compound on allergic immune responses remains unclear. In this study, we have examined the effects of OP on production of interleukin-4 (IL-4), a pro-inflammatory cytokine closely associated with allergic immune responses. OP significantly enhanced IL-4 production in antigen-primed T cells in a dose-dependent manner. Treatment with OP in vivo resulted in significant increase of IL-4 production in T cells and of IgE levels in sera of antigen-primed mice. Furthermore, OP enhanced the activation of IL-4 gene promoter in EL4 T cells transiently transfected with IL-4 promoter/reporter constructs, and the enhancing effect mapped to a region in the IL-4 promoter containing binding sites for nuclear factor of activated T cell (NF-AT). Activation of T cells by phorbol-12-myristate-13-acetate (PMA) resulted in markedly enhanced binding activities to the NF-AT site, which significantly increased upon addition of OP, indicating that the transcription factor NF-AT was involved in the enhancing effect of OP on IL-4 production. The enhancement of IL-4 production by OP was blocked by FK506, a calcineurin inhibitor, but not by the estrogen receptor (ER) antagonist ICI 182780. FK506 inhibited the NF-AT-DNA binding activity and IL-4 gene promoter activity enhanced by OP in a dose-dependent manner. These findings demonstrate that OP enhances IL-4 production in T cells via the stimulation of calcineurin-dependent NF-AT activation.
Collapse
Affiliation(s)
- Mi H Lee
- Immunology Laboratory, College of Pharmacy, Chonnam National University, Kwangju 500-757, South Korea
| | | | | |
Collapse
|
231
|
Jessop HL, Suswillo RFL, Rawlinson SCF, Zaman G, Lee K, Das-Gupta V, Pitsillides AA, Lanyon LE. Osteoblast-like cells from estrogen receptor alpha knockout mice have deficient responses to mechanical strain. J Bone Miner Res 2004; 19:938-46. [PMID: 15190886 DOI: 10.1359/jbmr.2004.19.6.938] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION In vivo, bones' osteogenic response to mechanical loading involves proliferation of surface osteoblasts. This response is replicated in vitro and involves ERK-mediated activation of the estrogen receptor (ER) alpha and upregulation of estrogen response element activity. This proliferative response can be blocked by selective estrogen receptor modulators and increased by transfection of additional ERalpha. MATERIALS AND METHODS We have now investigated the mechanisms of ER involvement in osteoblast-like cells' early responses to strain by comparing the responses of primary cultures of these cells derived from homozygous ERalpha knockout (ERKO) mice (ERalpha-/-) with those from their wildtype (ERalpha+/+) and heterozygous (ERalpha+/-) littermates and from ER/beta knockout (BERKO) mice (ERbeta+/+, ERbeta+/-, and ERbeta-/-). RESULTS Whereas ERalpha+/+, ERalpha+/-, ERbeta+/+, and ERbeta-/- cells proliferate in response to a single 10-minute period of cyclic strain, ERalpha-/- cells do not. Transfection of fully functional, but not mutant, ERalpha rescues the proliferative response to strain in these cells. The strain-related response of ERalpha-/- cells is also deficient in that they show no increased activity of an AP-I driven reporter vector and no strain-related increases in NO production. Their strain-related increase in prostacyclin production is retained. They proliferate in response to fibroblast growth factor-2 but not insulin-like growth factor (IGF)-I or IGF-II, showing the importance of ERalpha in the IGF axis and the ability of ERalpha-/- cells to proliferate normally in response to a mitogenic stimulus that does not require functional ERalpha. CONCLUSIONS These data indicate ERalpha's obligatory involvement in a number of early responses to mechanical strain in osteoblast-like cells, including those that result in proliferation. They support the hypothesis that reduction in ERalpha expression or activity after estrogen withdrawal results in a less osteogenic response to loading. This could be important in the etiology of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Helen L Jessop
- Department of Veterinary Basic Sciences, The Royal Veterinary College, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
232
|
Somjen D, Katzburg S, Sharon O, Kaye AM, Gayer B, Kohen F, Hendel D, Posner GH. Modulation of response to estrogens in cultured human female bone cells by a non-calcemic Vitamin D analog: changes in nuclear and membranal binding. J Steroid Biochem Mol Biol 2004; 89-90:393-5. [PMID: 15225807 DOI: 10.1016/j.jsbmb.2004.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Estradiol17beta (E2) and the phytoestrogens genistein (G), and daidzein (D) increase creatine kinase (CK) specific activity in primary cell cultures of human female to a greater extent in cells from pre-menopausal than post-menopausal women. Pretreatment with the non-calcemic analog of Vitamin D, JK 1624 F2-2 (JKF), upregulated this estrogenic response at all ages. In contrast, biochainin A (BA) and quercertin (Qu) increased CK with no age dependence or modulation by JKF pretreatment. Both ERalpha and ERbeta present in the cells were upregulated by pretreatment with JKF, as measured by Western blot analysis. Real time PCR showed no significant change in ERalpha mRNA but a marked decrease in ERbeta mRNA in both age groups after JKF treatment. Cells from both age groups had surface binding sites for E2, shown by assays using cell impermeable Europium labeled ovalbumin-E2 conjugate (Eu-Ov-E2). Binding of [3H]-E2 to intracellular E2 receptors (ERs) was similar in both age groups with differences in phytoestrogenic competition. JKF pretreatment increased nuclear but decreased membranal binding in both age groups. These results provide evidence for membranal, in addition to nuclear estrogen receptors which are differentially modulated by a Vitamin D analog.
Collapse
Affiliation(s)
- Dalia Somjen
- Institute of Endocrinology, Metabolism and Hypertension, Tel-Aviv Sourasky Medical Center, 6 Weizman St., Tel-Aviv 64239, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
233
|
Watson CS, Gametchu B. Proteins of multiple classes may participate in nongenomic steroid actions. Exp Biol Med (Maywood) 2004; 228:1272-81. [PMID: 14681543 PMCID: PMC1224708 DOI: 10.1177/153537020322801106] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Responses to steroids initiated from non-nuclear receptors impinge on a wide variety of cellular responses and utilize nearly all known signal transduction webs. While the mechanisms by which steroid receptors localize in the membrane are still unclear, it is apparent that this alternative localization allows steroid receptors to participate in a wide range of complex functions influencing cell proliferation, death, and differentiation. The central debate still remains the identity of the protein class or classes that mediate membrane-initiated (nongenomic) responses. The data thus far have supported several possibilities, including: nuclear steroid receptor-like forms in non-nuclear locations; other known (nonsteroid) membrane receptors or channels with additional steroid-binding sites; enzymes; transporters; receptors for serum steroid-binding proteins; unique and previously undescribed proteins; or chimeras of typical steroid receptor domains with other unique or known protein domains. Categorizing membrane steroid receptor proteins based exclusively on the actions of antagonists and agonists, without considering cell context and protein partnering issues, may mislead us into predicting more receptor subtypes than really exist. However, the plethora of signaling and functional outcomes may indicate the participation of more than one kind of steroid-binding protein. Resolving such unanswered questions will require future investigative focus on this alternative arm of steroid action, which is likely to yield as many therapeutic opportunities as have nuclear steroid mechanisms.
Collapse
Affiliation(s)
- Cheryl S Watson
- Department of Human Biological Chemistry and Genetics, University of Texas, Medical Branch, Galveston, Texas 77555, USA.
| | | |
Collapse
|
234
|
Soria B, Quesada I, Ropero AB, Pertusa JA, Martín F, Nadal A. Novel players in pancreatic islet signaling: from membrane receptors to nuclear channels. Diabetes 2004; 53 Suppl 1:S86-91. [PMID: 14749271 DOI: 10.2337/diabetes.53.2007.s86] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Glucose and other nutrients regulate many aspects of pancreatic islet physiology. This includes not only insulin release, but also insulin synthesis and storage and other aspects of beta-cell biology, including cell proliferation, apoptosis, differentiation, and gene expression. This implies that in addition to the well-described signals for insulin release, other intracellular signaling mechanisms are needed. Here we describe the role of global and local Ca(2+) signals in insulin release, the regulation of these signals by new membrane receptors, and the generation of nuclear Ca(2+) signals involved in gene expression. An integrated view of these pathways should improve the present description of the beta-cell biology and provide new targets for novel drugs.
Collapse
Affiliation(s)
- Bernat Soria
- Institute of Bioengineering, Miguel Hernandez University, Alicante, Spain.
| | | | | | | | | | | |
Collapse
|
235
|
Morales A, Díaz M, Ropero AB, Nadal A, Alonso R. Estradiol modulates acetylcholine-induced Ca2+ signals in LHRH-releasing GT1-7 cells through a membrane binding site. Eur J Neurosci 2003; 18:2505-14. [PMID: 14622151 DOI: 10.1046/j.1460-9568.2003.02997.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Estrogen regulation of the female reproductive axis involves the rapid inhibition (< 30 min) of luteinizing hormone-releasing hormone (LHRH) secretion from hypothalamic neurons. This fast time-course suggests interactions with potential plasma membrane binding sites that could result in short-term effects on LHRH neurons. Because LHRH release is calcium dependent, we have studied the acute effects of 17beta-estradiol (E2) and estradiol-peroxidase (E-HRP) on the elevations of intracellular calcium ([Ca2+]i) induced by acetylcholine (ACh) in LHRH-producing GT1-7 cells. Exposure to ACh (1-100 micro m) induced transient increases of [Ca2+]i, whereas pretreatment with E2 or E-HRP (10 nm) for 2 min reduced this response by 50-60%. The effect was specific for E2 as neither 17alpha-estradiol (1 micro m) nor the synthetic antiestrogens ICI182 780 (1 micro m) or tamoxifen (1 micro m) elicited any change on the ACh-induced Ca2+ signal. Both the latency of the effect and the response to the membrane impermeant conjugate suggested a membrane-mediated mechanism. Such membrane binding sites for E2 in GT1-7 cells were demonstrated by visualizing the binding of E-HRP and estradiol-BSA-fluorescein isothiocyanate (E-BSA-FITC) conjugates. Competition studies showed that E-HRP binding was blocked by preincubation with E2, but not with 17alpha-E2, ICI182 780, tamoxifen or progesterone, indicating that the plasma membrane binding site is highly specific for E2 and exhibits a pharmacological profile different from classical estrogen receptors. We conclude that ACh-induced increase in [Ca2+]i in GT1-7 cells is modulated acutely by physiological E2 concentrations in a manner which is compatible with the existence of an estrogen-specific membrane binding site.
Collapse
Affiliation(s)
- Araceli Morales
- Department of Physiology, University of La Laguna, Tenerife, Spain
| | | | | | | | | |
Collapse
|
236
|
Booth EA, Marchesi M, Kilbourne EJ, Lucchesi BR. 17Beta-estradiol as a receptor-mediated cardioprotective agent. J Pharmacol Exp Ther 2003; 307:395-401. [PMID: 12893838 DOI: 10.1124/jpet.103.054205] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cardiac tissue that undergoes an ischemic episode exhibits irreversible alterations that become more extensive upon reperfusion. Estrogen treatment has been reported to protect against reperfusion injury, but the mechanism remains unknown. The cardioprotective effects of 17beta-estradiol, a biologically active form of the hormone, and 17alpha-estradiol were assessed in an in vivo occlusion-reperfusion model. Anesthetized, ovariectomized rabbits were administered 17beta-estradiol (20 microg), 17alpha-estradiol (1 mg), or vehicle intravenously 30 min before a 30-min occlusion of the left anterior descending (LAD) coronary artery followed by 4 h of reperfusion. Infarct size as a percentage of area at risk decreased in the 17beta-estradiol-treated group (18.8 +/- 1.7) compared with 17alpha-estradiol (41.9 +/- 4.8; P < 0.01) or vehicle groups (48 +/- 5.5; P < 0.001). Similar results were obtained when infarct size was expressed as a percentage of total left ventricle. The second objective of the study was to assess fulvestrant (Faslodex, ICI 182,780), an estrogen receptor antagonist, for its effects on infarct size in ovariectomized female rabbits treated with 17beta-estradiol. ICI 182,780 was administered intravenously 1 h before the administration of 17beta-estradiol (20 microg) or vehicle. The hearts were subjected to 30-min LAD coronary artery occlusion and 4 h of reperfusion. Pretreatment with ICI 182,780 significantly limited the infarct size sparing effect of 17beta-estradiol when expressed as a percentage of the risk region (53.0 +/- 5.0). The results indicate that 17beta-estradiol protects the heart against ischemia-reperfusion injury and that the observed cardioprotection is mediated by the estrogen receptor.
Collapse
Affiliation(s)
- Erin A Booth
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632, USA
| | | | | | | |
Collapse
|
237
|
Abstract
Oestrogen is important for the development of neuroendocrine centres and other neural networks including limbic and motor systems. Later in adulthood, oestrogen regulates the functional performance of different neural systems and is presumably implicated in the modulation of cognitive efficiency. Although still a matter of controversial discussion, clinical and experimental studies point at a potential neuroprotective role of oestrogen. Concerning the concept of cellular oestrogen action, it is undisputed that it comprises the binding and activation of nuclear receptors. The last decades have, however, immensely broadened the spectrum of steroid signalling within a cell. Novel steroid-activated intracellular signalling mechanisms were described which are usually termed 'non-classical' or 'non-genomic'. The brain appears to be a rich source of this new mode of oestrogen action. Studies from the past years have pinpointed non-classical oestrogen effects in many CNS regions. All available data support the view that non-classical oestrogen action requires interactions with putative membrane binding sites/receptors. In this article, we aim at compiling the most recent findings on the nature and identity of membrane oestrogen receptors with respect to the brain. We also attempt to turn readers attention to the coupling of these 'novel' receptors to distinct intracellular signalling pathways.
Collapse
Affiliation(s)
- Cordian Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Ulm, Germany.
| | | | | |
Collapse
|
238
|
Estrogen receptor beta mediates rapid estrogen actions on gonadotropin-releasing hormone neurons in vivo. J Neurosci 2003. [PMID: 12843281 DOI: 10.1523/jneurosci.23-13-05771.2003] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The gonadal steroid estrogen exerts an important modulatory influence on the activity of multiple neuronal networks. In addition to classical genomic mechanisms of action, estrogen also exerts poorly understood rapid, nongenomic effects on neurons. To examine whether estrogen may exert rapid actions on intracellular signaling within gonadotropin-releasing hormone (GnRH) neurons in vivo,we examined the phosphorylation status of cAMP response element-binding protein (CREB) in these cells after the administration of 17-beta-estradiol to ovariectomized (OVX) mice. The percentage of GnRH neurons expressing phosphorylated CREB was increased more than sixfold (p < 0.05) in a time- and dose-dependent manner by estrogen, with the increase first observed 15 min after estrogen administration. A series of in vitro studies demonstrated that estrogen acted directly on native GnRH neurons to phosphorylate CREB, but that estrogen conjugated to bovine serum albumin was without effect. The role of classical estrogen receptors (ERs) was evaluated using ER knock-out mice in vivo. The effect of estrogen on CREB phosphorylation in GnRH neurons was normal in ERalpha knock-out mice but completely absent in ERbeta knock-out mice. Finally, studies in intact female mice revealed levels of CREB phosphorylation within GnRH neurons that were equivalent to those of estrogen-treated OVX mice. These observations demonstrate that ERbeta mediates the rapid, direct effects of estrogen on the GnRH neuronal phenotype, and that these actions persist under physiological conditions. They also provide the first evidence for a role of ERbeta in nongenomic estrogen signaling within the brain in vivo.
Collapse
|
239
|
Nativelle-Serpentini C, Richard S, Séralini GE, Sourdaine P. Aromatase activity modulation by lindane and bisphenol-A in human placental JEG-3 and transfected kidney E293 cells. Toxicol In Vitro 2003; 17:413-22. [PMID: 12849724 DOI: 10.1016/s0887-2333(03)00046-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Aromatase is the cytochrome P-450 involved in converting androgens to estrogens. The cytochrome P-450 family plays a central role in the oxidative metabolism of compounds including environmental pollutants. Since lindane and bisphenol-A (BPA) are two well-characterized endocrine disruptors that have been detected in animals and humans, it was important to learn whether they could affect aromatase activity and consequently estrogen biosynthesis. The present study investigates the effects of BPA and lindane on cytotoxicity, aromatase activity and mRNA levels in human placental JEG-3 cells and transfected human embryonal kidney 293 cells. Both cell lines were exposed to increasing concentrations of lindane (25, 50 and 75 microM) and bisphenol-A (25, 50 and 100 microM) over different time periods (10 min-18 h). As a result, none of these concentrations showed cytotoxicity. After short pre-incubation times (10 min-6 h), aromatase activity was enhanced by both compounds. Longer time incubation (18 h), however, produced dose-related inhibition. Lindane and BPA had no significant effects on CYP19 mRNA levels. Therefore, lindane and BPA modulate aromatase activity suggesting an interaction with the cytochrome P-450 aromatase. This study highlights the endocrine-modulating properties of lindane and bisphenol-A.
Collapse
Affiliation(s)
- C Nativelle-Serpentini
- IBBA - Laboratoire de Biochimie et Biologie Moléculaire - EA 2608 - Université de Caen, Esplanade de la Paix - 14032, Caen cedex, France
| | | | | | | |
Collapse
|
240
|
Chiyo T, Yamazaki T, Aoshika K, Kominami S, Ohta Y. Corticosterone enhances adrenocorticotropin-induced calcium signals in bovine adrenocortical cells. Endocrinology 2003; 144:3376-81. [PMID: 12865316 DOI: 10.1210/en.2002-221126] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The rapid effects of steroid hormones on Ca(2+) signals have been examined in bovine adrenocortical cells. Among the steroid molecules tested, only corticosterone rapidly stimulated Ca(2+) signals upon addition of ACTH, although corticosterone alone did not induce Ca(2+) signals. Corticosterone also enhanced steroidogenesis induced by ACTH. The enhancement of ACTH-induced Ca(2+) signals was also observed with membrane-impermeable corticosterone conjugated to BSA and was not inhibited by cycloheximide. In addition, corticosterone did not enhance Ca(2+) signals induced by ATP or angiotensin II. These results suggest that corticosterone selectively stimulates ACTH-induced Ca(2+) signals in a nongenomic way by acting on a target in the plasma membrane. Furthermore, the supernatants of cells incubated with ACTH or ATP enhanced Ca(2+) signals, suggesting that steroids produced by such treatment act in an autocrine fashion. Consistent with this idea, these effects were inhibited by inhibitors of steroidogenesis (aminoglutethimide or metyrapone). These results show that steroid molecules synthesized in adrenocortical cells facilitate ACTH-induced Ca(2+) signals. Taken together, corticosterone secreted from adrenocortical cells activates ACTH-induced Ca(2+) signals and steroidogenesis by nongenomic means.
Collapse
Affiliation(s)
- Tomoko Chiyo
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Nakacho, Koganei, Tokyo, 184-8588, Japan
| | | | | | | | | |
Collapse
|
241
|
Abrahám IM, Han SK, Todman MG, Korach KS, Herbison AE. Estrogen receptor beta mediates rapid estrogen actions on gonadotropin-releasing hormone neurons in vivo. J Neurosci 2003; 23:5771-7. [PMID: 12843281 PMCID: PMC6741236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
The gonadal steroid estrogen exerts an important modulatory influence on the activity of multiple neuronal networks. In addition to classical genomic mechanisms of action, estrogen also exerts poorly understood rapid, nongenomic effects on neurons. To examine whether estrogen may exert rapid actions on intracellular signaling within gonadotropin-releasing hormone (GnRH) neurons in vivo,we examined the phosphorylation status of cAMP response element-binding protein (CREB) in these cells after the administration of 17-beta-estradiol to ovariectomized (OVX) mice. The percentage of GnRH neurons expressing phosphorylated CREB was increased more than sixfold (p < 0.05) in a time- and dose-dependent manner by estrogen, with the increase first observed 15 min after estrogen administration. A series of in vitro studies demonstrated that estrogen acted directly on native GnRH neurons to phosphorylate CREB, but that estrogen conjugated to bovine serum albumin was without effect. The role of classical estrogen receptors (ERs) was evaluated using ER knock-out mice in vivo. The effect of estrogen on CREB phosphorylation in GnRH neurons was normal in ERalpha knock-out mice but completely absent in ERbeta knock-out mice. Finally, studies in intact female mice revealed levels of CREB phosphorylation within GnRH neurons that were equivalent to those of estrogen-treated OVX mice. These observations demonstrate that ERbeta mediates the rapid, direct effects of estrogen on the GnRH neuronal phenotype, and that these actions persist under physiological conditions. They also provide the first evidence for a role of ERbeta in nongenomic estrogen signaling within the brain in vivo.
Collapse
Affiliation(s)
- István M Abrahám
- Laboratory of Neuroendocrinology, The Babraham Institute, Cambridge CB2 4AT, United Kingdom
| | | | | | | | | |
Collapse
|
242
|
Losel RM, Falkenstein E, Feuring M, Schultz A, Tillmann HC, Rossol-Haseroth K, Wehling M. Nongenomic steroid action: controversies, questions, and answers. Physiol Rev 2003; 83:965-1016. [PMID: 12843413 DOI: 10.1152/physrev.00003.2003] [Citation(s) in RCA: 393] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Steroids may exert their action in living cells by several ways: 1). the well-known genomic pathway, involving hormone binding to cytosolic (classic) receptors and subsequent modulation of gene expression followed by protein synthesis. 2). Alternatively, pathways are operating that do not act on the genome, therefore indicating nongenomic action. Although it is comparatively easy to confirm the nongenomic nature of a particular phenomenon observed, e.g., by using inhibitors of transcription or translation, considerable controversy exists about the identity of receptors that mediate these responses. Many different approaches have been employed to answer this question, including pharmacology, knock-out animals, and numerous biochemical studies. Evidence is presented for and against both the participation of classic receptors, or proteins closely related to them, as well as for the involvement of yet poorly understood, novel membrane steroid receptors. In addition, clinical implications for a wide array of nongenomic steroid actions are outlined.
Collapse
Affiliation(s)
- Ralf M Losel
- Institut für klinische Pharmakologie, Klinikum Mannheim, Theodor-Kutzer-Ufer, D-68167 Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
243
|
Marin R, Guerra B, Morales A, Díaz M, Alonso R. An oestrogen membrane receptor participates in estradiol actions for the prevention of amyloid-beta peptide1-40-induced toxicity in septal-derived cholinergic SN56 cells. J Neurochem 2003; 85:1180-9. [PMID: 12753077 DOI: 10.1046/j.1471-4159.2003.01767.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although oestrogen [17 beta-estradiol (E2)]-related neuroprotection has been demonstrated in different models, the involvement of non-classical oestrogen receptors (ERs) remains unexplored. Using the SN56 cholinergic cell line, we present evidence indicating that an ER associated with the plasma membrane participates in oestrogen-dependent inhibition of cell death induced by amyloid-beta peptide (A beta) toxicity. Similarly to E2 alone, a 15-min exposure to estradiol-horseradish peroxidase (E-HRP) significantly reduced A beta-induced cell death. This effect was decreased by the ER antagonist ICI 182,780 as well as by MC-20 antibody directed to a region neighbouring the ligand-binding domain of ER alpha. Using confocal microscopy on unpermeabilized SN56 cells exposed to MC-20 antibody, we identified a protein at the plasma membrane level. Western blot analysis of purified SN56 cell membrane fractions using MC-20 antibody revealed the presence of one band with the same electrophoretic mobility as intracellular ER alpha. Using conjugated forms of the steroid, E-HRP and E2 conjugated to bovine serum albumin-FITC, we demonstrated by confocal microscopy that SN56 cells contain surface binding sites for E2. Binding of both conjugates was blocked by pre-incubation with E2 and decreased by either ICI 182,780 or MC-20 antibody in a concentration-dependent manner. Thus, a membrane-related ER that shares some structural homologies with ER alpha may participate in oestrogen-mediated neuroprotection.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Physiology, University of La Laguna, School of Medicine, Sta. Cruz de Tenerife, Spain.
| | | | | | | | | |
Collapse
|
244
|
Abstract
Fear is an adaptive component of the acute "stress" response to potentially-dangerous (external and internal) stimuli which threaten to perturb homeostasis. However, when disproportional in intensity, chronic and/or irreversible, or not associated with any genuine risk, it may be symptomatic of a debilitating anxious state: for example, social phobia, panic attacks or generalized anxiety disorder. In view of the importance of guaranteeing an appropriate emotional response to aversive events, it is not surprising that a diversity of mechanisms are involved in the induction and inhibition of anxious states. Apart from conventional neurotransmitters, such as monoamines, gamma-amino-butyric acid (GABA) and glutamate, many other modulators have been implicated, including: adenosine, cannabinoids, numerous neuropeptides, hormones, neurotrophins, cytokines and several cellular mediators. Accordingly, though benzodiazepines (which reinforce transmission at GABA(A) receptors), serotonin (5-HT)(1A) receptor agonists and 5-HT reuptake inhibitors are currently the principle drugs employed in the management of anxiety disorders, there is considerable scope for the development of alternative therapies. In addition to cellular, anatomical and neurochemical strategies, behavioral models are indispensable for the characterization of anxious states and their modulation. Amongst diverse paradigms, conflict procedures--in which subjects experience opposing impulses of desire and fear--are of especial conceptual and therapeutic pertinence. For example, in the Vogel Conflict Test (VCT), the ability of drugs to release punishment-suppressed drinking behavior is evaluated. In reviewing the neurobiology of anxious states, the present article focuses in particular upon: the multifarious and complex roles of individual modulators, often as a function of the specific receptor type and neuronal substrate involved in their actions; novel targets for the management of anxiety disorders; the influence of neurotransmitters and other agents upon performance in the VCT; data acquired from complementary pharmacological and genetic strategies and, finally, several open questions likely to orientate future experimental- and clinical-research. In view of the recent proliferation of mechanisms implicated in the pathogenesis, modulation and, potentially, treatment of anxiety disorders, this is an opportune moment to survey their functional and pathophysiological significance, and to assess their influence upon performance in the VCT and other models of potential anxiolytic properties.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, Centre de Rescherches de Croissy, Institut de Recherches (IDR) Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, Paris, France.
| |
Collapse
|
245
|
Estrada-Camarena E, Fernández-Guasti A, López-Rubalcava C. Antidepressant-like effect of different estrogenic compounds in the forced swimming test. Neuropsychopharmacology 2003; 28:830-8. [PMID: 12637949 DOI: 10.1038/sj.npp.1300097] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The present study evaluated the possible antidepressant-like action of the natural estrogen 17beta-estradiol (E(2), 2.5-10 microg/rat), the synthetic steroidal estrogen ethinyl-estradiol (EE(2), 1.25-10.0 microg/rat), and the nonsteroidal synthetic estrogen, diethyl-stilbestrol (DES, 0.25-1.0 mg/rat) in ovariectomized adult female Wistar rats using the forced swimming test (FST). The behavioral profile induced by the estrogens was compared with that induced by the antidepressants fluoxetine (FLX, 2.5-10 mg/kg) and desipramine (DMI, 2.5-10 mg/kg). In addition, the temporal course of the antidepressant-like action of the estrogenic compounds was analyzed. FLX and DMI induced an antidepressant-like effect characterized by a reduced immobility and increased swimming for FLX and decreased immobility and increased climbing for DMI. Both E(2) and EE(2) produced a decrease in immobility and an increase in swimming, suggesting an antidepressant-like action. DES did not affect the responses in this animal model of depression at any dose tested. The time course analysis of the actions of E(2) (10 microg/rat) and EE(2) (5 microg/rat) showed that both compounds induced an antidepressant-like effect observed 1 h after their injection lasting for 2-3 days.
Collapse
Affiliation(s)
- Erika Estrada-Camarena
- Depto de Farmacobiología CINVESTAV-IPN, Calzada de los Tenorios 235, Col Granjas Coapa, Deleg Tlalpan, CP 14330 México DF, México.
| | | | | |
Collapse
|
246
|
Wade CB, Dorsa DM. Estrogen activation of cyclic adenosine 5'-monophosphate response element-mediated transcription requires the extracellularly regulated kinase/mitogen-activated protein kinase pathway. Endocrinology 2003; 144:832-8. [PMID: 12586759 DOI: 10.1210/en.2002-220899] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ability of estrogen to rapidly initiate a variety of signal transduction cascades is increasingly recognized as playing an important role in a number of tissue-specific transcriptional actions of the hormone. In vivo, estrogen rapidly elicits phosphorylation of cAMP response element-binding protein (CREB). We have previously shown that both ER alpha and ER beta are capable of activating the MAPK pathway in response to a low dose of 17beta-estradiol. In the present study, the ability of estrogen to act through both ER alpha and ER beta to increase CREB phosphorylation was evaluated in an immortalized hippocampal cell line stably expressing either receptor. Estrogen treatment promoted rapid CREB phosphorylation, reaching a maximum by 15 min. This activation is completely blocked by the antiestrogen ICI 182,780, suggesting an estrogen receptor-dependent mechanism. The addition of the mitogen/ERK kinase-1 inhibitor, PD98059, also blocked the ability of estrogen to signal to CREB phosphorylation. Estrogen also caused an increase in p90Rsk activity, a critical mediator of MAPK effects. Surprisingly, blockade of the protein kinase A pathway in cells treated with estrogen did not affect estrogen-mediated CREB phosphorylation. Thus, MAPK and p90Rsk appear to be the primary mediators of estrogen-induced gene transcription through ER alpha and ER beta.
Collapse
Affiliation(s)
- Christian B Wade
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | | |
Collapse
|
247
|
Flores-Delgado G, Anderson KD, Warburton D. Nongenomic estrogen action regulates tyrosine phosphatase activity and tuberin stability. Mol Cell Endocrinol 2003; 199:143-51. [PMID: 12581886 DOI: 10.1016/s0303-7207(02)00288-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Estrogen action and tuberin function has been suggested to play a crucial role in the proliferation of lung smooth muscle-like cells and/or myofibroblasts in pulmonary lymphangioleiomyomatosis (LAM). Tuberin is a tumor suppressor phosphoprotein, which also regulates fluid phase endocytosis. Its activity, turnover and complex association with hamartin depends on its phosphorylation status. We have recently reported that nongenomic estrogen action regulates the phosphorylation status of several cytoplasmic proteins. Herein, we demonstrate that estrogen increases tyrosine phosphatase activity, which can be abrogated by antiestrogen ICI 182780 and tyrosine phosphatase inhibitor bpV(phen), but not by the protein synthesis inhibitor cyclohexamide. Furthermore, we show that estrogen transiently enhances the turnover of tuberin, which follows an inverse pattern to that observed for tyrosine phosphatase and endocytosis activity. We showed that tuberin phosphorylation protects it from degradation and induces its accumulation in female human lung fibroblasts and myofibroblasts. Our results suggest that nongenomic estrogen action induces tyrosine phosphatase activity that regulates stability of tyrosine phosphorylated proteins, including tuberin, which may play a crucial role in cellular specific functions such as endocytosis.
Collapse
Affiliation(s)
- Guillermo Flores-Delgado
- Department of Surgery and the Developmental Biology Program, Childrens Hospital Los Angeles Research Institute, University of Southern California, 4650, Sunset Blvd. MS#35, 90027, USA.
| | | | | |
Collapse
|
248
|
Doolan CM, Harvey BJ. A Galphas protein-coupled membrane receptor, distinct from the classical oestrogen receptor, transduces rapid effects of oestradiol on [Ca2+]i in female rat distal colon. Mol Cell Endocrinol 2003; 199:87-103. [PMID: 12581882 DOI: 10.1016/s0303-7207(02)00303-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We examined the hypothesis whether rapid non-genomic effects of oestradiol (E2) on [Ca(2+)](i) are mediated via a membrane-located oestrogen receptor (ER) and further elucidated the signalling pathways involved in rapid non-genomic effects of E2 on [Ca(2+)](i) in distal colonic crypts. Basal [Ca(2+)](i) was significantly increased, within minutes, in response to physiological concentrations of E2. Oestradiol linked to bovine serum albumin (E2-BSA), which renders the E2 membrane impermeable, rapidly increased [Ca(2+)](i) suggesting mediation by a membrane surface receptor. A classical ER is not involved however, as no inhibition of either the E2 or E2-BSA [Ca(2+)](i) response was seen in the presence of the classical ER antagonist ICI 182,780. Treatment with the Galphas inhibitor cholera toxin abolished both E2 and E2-BSA induced Ca(2+) increases. In contrast, treatment with pertussis toxin, an inhibitor of Galphai and Galphao, had no inhibitory effect. Following subsequent additions of E2 and E2-BSA, no further increases in [Ca(2+)](i) were observed, indicating receptor desensitisation. The E2-induced increase in [Ca(2+)](i) was completely abolished by the PKCdelta-specific inhibitor rottlerin, whereas Go6976, an inhibitor of Ca(2+)-sensitive PKC isoforms, was without inhibitory effect. The phospholipase A2 antagonist, quinacrine, and the COX1 inhibitor, indomethacin, abolished the E2-induced increase in [Ca(2+)](i). MAP kinase activation is not involved in rapid stimulatory effects of E2 on [Ca(2+)](i) as the specific inhibitor PD98059 did not inhibit the E2 response. These results demonstrate that rapid E2-induced stimulation of [Ca(2+)](i), in femal rat distal colonic crypts, occurs via a CTx-sensitive Galphas-coupled membrane receptor distinct from the classical ER. PKCdelta and fatty acids are involved in the E2 signalling pathway. In contrast, PKCalpha and MAP kinase are not required.
Collapse
Affiliation(s)
- Christina M Doolan
- Department of Physiology, Biosciences Institute, University College Cork, Ireland.
| | | |
Collapse
|
249
|
Amstislavsky SY, Kizilova EA, Eroschenko VP, Amstislavksy SY. Preimplantation mouse embryo development as a target of the pesticide methoxychlor. Reprod Toxicol 2003; 17:79-86. [PMID: 12507662 DOI: 10.1016/s0890-6238(02)00095-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Effects of methoxychlor (MXC) and estradiol-17beta (E) were studied in mouse preimplantation embryos. Pregnant mice received s.c. injections of sesame oil only, 10 microg E, or 0.5 mg purified (95%) MXC on Days 2-4 of pregnancy (plug = Day 1). Another group received a single dose of 2.5 microg E on Day 2 only. Based on the average weight of pregnant females, 10 microg of estradiol was equivalent to 0.33 mg/kg of bw, 2.5 microg of estradiol was equivalent to 0.082 mg/kg of bw, and the 0.5-mg dose of MXC was equivalent to 16.5 mg/kg of bw. All embryos were collected for analyses on Day 4. MXC and both estradiol-17beta doses suppressed embryonic development to blastocyst, decreased embryo cell numbers, and caused abnormal blastocyst formation. The high estradiol-17beta dose significantly increased the percent degenerating embryos and caused a tube-locking effect, with retention of embryos in the oviduct. In contrast to estradiol-17beta, MXC at the dose used in this study did not alter tubal transport of embryos. Also in contrast to estradiol-17beta, MXC increased the percentage of nuclear fragmentation and micronuclei. In preimplantation embryos, MXC and estradiol-17beta both suppressed embryo development. MXC effects were, however, different from those of estradiol-17beta, indicating a difference in mechanism of action, possibly due to cytotoxicity and induction of apoptosis.
Collapse
Affiliation(s)
- Sergei Y Amstislavsky
- Institute of Cytology and Genetics, Russian Academy of Sciences, Siberian Division, Novosibirsk State University, Novosibirsk, Russia
| | | | | | | |
Collapse
|
250
|
Frigo DE, Burow ME, Mitchell KA, Chiang TC, McLachlan JA. DDT and its metabolites alter gene expression in human uterine cell lines through estrogen receptor-independent mechanisms. ENVIRONMENTAL HEALTH PERSPECTIVES 2002; 110:1239-45. [PMID: 12460804 PMCID: PMC1241112 DOI: 10.1289/ehp.021101239] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Endocrine-disrupting organochlorines, such as the pesticide dichlorodiphenyltrichloroethane (DDT), bind to and activate estrogen receptors (ERs), thereby eliciting estrogen-like effects. Although ERs function predominantly through activation of transcription via estrogen-responsive elements, both ERs, alpha and ss, can interact with various transcription factors such as activator protein-1 (AP-1). Additionally, estrogens may regulate early signaling events, suggesting that the biological effects of environmental estrogens may not be mediated through classic ER (alpha and ss) activity alone. We hypothesized that known environmental estrogens, such as DDT and its metabolites, activate AP-1-mediated gene transactivation through both ER-dependent and ER-independent means. Using two Ishikawa human endometrial adenocarcinoma cell line variants that we confirmed to be estrogen responsive [Ishikawa(+)] and estrogen unresponsive [Ishikawa(-)], we generated stably transfected AP-1 luciferase cell lines to identify the role of an estrogen-responsive mechanism in AP-1-mediated gene expression by various stimuli. Our results demonstrate that DDT and dichlorodiphenyldichloroethane (DDD) were the most potent activators of AP-1 activity; 2,2-bis(p-chlorophenyl) acetic acid failed to activate. Although stimulated in both Ishikawa(+) and Ishikawa(-) cells by DDT and its congeners, AP-1 activation was more pronounced in the estrogen-unresponsive Ishikawa(-) cells. In addition, DDT, DDD, and dichlorodiphenyldichloroethylene (DDE) could also stimulate AP-1 activity in the estrogen-unresponsive human embryonic kidney 293 cells using a different promoter context. Thus, our data demonstrate that DDT and its metabolites activate the AP-1 transcription factor independent of ER (alpha or ss) status.
Collapse
Affiliation(s)
- Daniel E Frigo
- Molecular and Cellular Biology Program, Tulane University Health Science Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|