301
|
Abstract
BACKGROUND While the androgen-dependence of the prostate gland has long been accepted, the participation of estrogen, mediated via the stroma in the elicitation of benign prostatic hyperplasia (BPH), has only recently been recognized. Its mode of action is still uncertain. METHODS This review first outlines the regulation of gene expression via hormones, growth factors, and other ligands in the coordination of cell growth, differentiation, and function. Focus is next directed to factors particularly involved in phosphorylation of estrogen receptors. Then, the access of sex steroids, especially of estrogen to the cell and to the transduction machinery, is described, preparatory to examining the hypotheses by which this access causes the process of BPH to occur. RESULTS It becomes clear that the necessary phosphorylative activities which transmit signals to nuclear receptors and thence transcription of target genes can be performed by steroids or mimicked by proxy molecules and by cross-talk between discrete pathways. The character and concentration of the available estrogen are determined by the extent of its biosynthesis, its penetration of the cell, and its subsequent metabolism. In addition, the estrogen affects its own access through stimulation of facilitating peptide hormones, prolactin, and sex hormone-binding globulin. Finally, the induction of BPH is shown to be determined by the androgen/estrogen ratio and the change in stromal/epithelial balance accompanying aging. CONCLUSIONS Despite a growing knowledge of hormone levels, metabolism, and activities in the prostate, and the variety of processes and factors they affect, our explanation of BPH is still fanciful.
Collapse
Affiliation(s)
- W E Farnsworth
- Department of Urology, Northwestern University Medical School, Bloomingdale, Illinois 60108-2833, USA.
| |
Collapse
|
302
|
Ropero AB, Fuentes E, Rovira JM, Ripoll C, Soria B, Nadal A. Non-genomic actions of 17beta-oestradiol in mouse pancreatic beta-cells are mediated by a cGMP-dependent protein kinase. J Physiol 1999; 521 Pt 2:397-407. [PMID: 10581311 PMCID: PMC2269678 DOI: 10.1111/j.1469-7793.1999.00397.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/1999] [Accepted: 10/04/1999] [Indexed: 11/28/2022] Open
Abstract
1. Intracellular calcium concentration ([Ca2+]i) was measured in mouse whole islets of Langerhans using the calcium-sensitive fluorescent dye Indo-1. 2. Application of physiological concentrations of 17beta-oestradiol in the presence of a stimulatory glucose concentration (8 mM) potentiated the [Ca2+]i signal in 83 % of islets tested. Potentiation was manifested as either an increase in the frequency or duration of [Ca2+]i oscillations. 3. The effects caused by 17beta-oestradiol were mimicked by the cyclic nucleotide analogues 8-bromoguanosine-3',5'-cyclic monophosphate (8-Br-cGMP) and 8-bromoadenosine-3',5'-cyclic monophosphate (8-Br-cAMP). 4. Direct measurements of both cyclic nucleotides demonstrated that nanomolar concentrations of 17beta-oestradiol in the presence of 8 mM glucose increased cGMP levels, yet cAMP levels were unchanged. The increment in cGMP was similar to that induced by 11 mM glucose. 5. Patch-clamp recording in intact cells showed that 8-Br-cGMP reproduced the inhibitory action of 17beta-oestradiol on ATP-sensitive K+ (KATP) channel activity. This was not a membrane-bound effect since it could not be observed in excised patches. 6. The action of 17beta-oestradiol on KATP channel activity was not modified by the specific inhibitor of soluble guanylate cyclase (sGC) LY 83583. This result indicates a likely involvement of a membrane guanylate cyclase (mGC). 7. The rapid decrease in KATP channel activity elicited by 17beta-oestradiol was greatly reduced using Rp-8-pCPT-cGMPS, a specific blocker of cGMP-dependent protein kinase (PKG). Conversely, Rp-cAMPS, which inhibits cAMP-dependent protein kinase (PKA), had little effect. 8. The results presented here indicate that rapid, non-genomic effects of 17beta-oestradiol after interaction with its binding site at the plasma membrane of pancreatic beta-cells is a cGMP-dependent phosphorylation process.
Collapse
Affiliation(s)
- A B Ropero
- Institute of Bioengineering and Department of Physiology, Miguel Hernández University, San Juan Campus, Alicante, Spain
| | | | | | | | | | | |
Collapse
|
303
|
Abstract
Estrogen exerts long-term effects in almost every cell through regulation of gene transcription. However, it has been known for some time that estrogen can rapidly alter neuronal firing within seconds, indicating that some cellular effects of estrogen could occur via non-genomic mechanisms. G-protein-coupled receptors (GPCRs) are the largest class of membrane-bound receptors, and it appears that many of the rapid effects mediated by estrogen could involve changes in GPCR-effector system coupling in excitable cells within the reproductive axis.
Collapse
|
304
|
Abstract
The existence of an estrogen receptor (ER) on the plasma membrane has been supported by data emerging from numerous laboratories over the past 20 years. However, this receptor has not yet been isolated. Original reports of a cell membrane protein that could bind and rapidly respond to 17beta-estradiol (E2) were supported by evidence that a putative membrane receptor could effect a variety of signal transduction events. Recent studies have shown that the nongenomic actions of E2 can be mediated through the plasma membrane ER.
Collapse
Affiliation(s)
- ER Levin
- aDivision of Endocrinology, Veterans Affairs Medical Center, Long Beach, CA 90822, USA
| |
Collapse
|
305
|
Abstract
The effect of estrogen on plasma membrane potential of isolated avian osteoclasts was examined through the use of a fluorescent potential-sensitive dye, bis-(1,3-dibutylbarbiturate) trimethine oxonol, also known as bis-oxonol. A decrease in potential was observed within seconds of addition of 17beta-estradiol. Ouabain, a specific Na+K+-ATPase inhibitor, and BaCl2, an inhibitor of the inwardly rectifying K+ channel, blocked the estrogen response. Verapamil and lanthanum chloride (LaCl3), inhibitors of inward Ca2+ channels, and 4'4-diisothiocyanatostilbene-2'2-disulfonic acid (DIDS), an inhibitor of Cl- channels, did not affect the depolarization. Herbimycin A, a tyrosine kinase inhibitor, also had no effect on the decreased membrane potential. These data provide evidence which suggests that estrogen regulates osteoclasts through ion channel activities. The change in K+ channel activity was observed within seconds of addition of 17beta-estradiol, indicating an action at the level of the plasma membrane.
Collapse
Affiliation(s)
- K D Brubaker
- Department of Biochemistry, The Pennsylvania State University, University Park, Pennsylvania, USA
| | | |
Collapse
|
306
|
Watson CS, Campbell CH, Gametchu B. Membrane oestrogen receptors on rat pituitary tumour cells: immuno-identification and responses to oestradiol and xenoestrogens. Exp Physiol 1999; 84:1013-22. [PMID: 10564698 PMCID: PMC1931420 DOI: 10.1111/j.1469-445x.1999.01903.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Our laboratory has identified plasma membrane oestrogen receptors on a GH3/B6 rat pituitary tumour cell line and several sublines which produce rapid (within minutes), non-genomic responses to oestrogens. Oestrogen receptors have been identified by their binding to nine different antibodies (Abs) which together recognize at least seven epitopes on the oestrogen receptor-alpha. GH3/B6/F10 cells, a membrane oestrogen receptor-enriched subline, elevate intracellular calcium levels in response to 10 nM oestradiol. Prolactin release in these cells is triggered by both 1 pM and 1 nM oestradiol and diethylstilbestrol (DES). A membrane oestrogen receptor-alpha immunocytochemical signal rapidly disappears (at 3 min) and reappears (at 12-15 min) when 1 nM oestradiol, 10 nM diethylstilbestrol, or 10 nM nonylphenol is applied to the cells. This suggests that both oestrogens and xenoestrogens can utilize this alternative pathway for oestrogenic action. Xenoestrogens, which have so far shown weak effects in genomic assay systems, should now be retested for activity in eliciting membrane-initiated oestrogenic responses.
Collapse
Affiliation(s)
- C S Watson
- Human Biological Chemistry & Genetics Department, University of Texas Medical Branch, Galveston, TX 77555-0645, USA.
| | | | | |
Collapse
|
307
|
Valverde MA, Rojas P, Amigo J, Cosmelli D, Orio P, Bahamonde MI, Mann GE, Vergara C, Latorre R. Acute activation of Maxi-K channels (hSlo) by estradiol binding to the beta subunit. Science 1999; 285:1929-31. [PMID: 10489376 DOI: 10.1126/science.285.5435.1929] [Citation(s) in RCA: 401] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Maxi-K channels consist of a pore-forming alpha subunit and a regulatory beta subunit, which confers the channel with a higher Ca(2+) sensitivity. Estradiol bound to the beta subunit and activated the Maxi-K channel (hSlo) only when both alpha and beta subunits were present. This activation was independent of the generation of intracellular signals and could be triggered by estradiol conjugated to a membrane-impenetrable carrier protein. This study documents the direct interaction of a hormone with a voltage-gated channel subunit and provides the molecular mechanism for the modulation of vascular smooth muscle Maxi-K channels by estrogens.
Collapse
Affiliation(s)
- M A Valverde
- Departament de Ciències Experimentals i de la Salut, Universidad Pompeu Fabra, C/Doctor Aiguader 80, 08003 Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
308
|
Kim HP, Lee JY, Jeong JK, Bae SW, Lee HK, Jo I. Nongenomic stimulation of nitric oxide release by estrogen is mediated by estrogen receptor alpha localized in caveolae. Biochem Biophys Res Commun 1999; 263:257-62. [PMID: 10486286 DOI: 10.1006/bbrc.1999.1348] [Citation(s) in RCA: 223] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acute administration of 17beta-estradiol (E(2)) exerts antiatherosclerotic effects in healthy postmenopausal women. The vasoprotective action of E(2) may be partly accounted for by a rapid increase in nitric oxide (NO) levels in endothelial cells (ECs). However, the signaling mechanisms producing this rise are unknown. In an attempt to address the short-term effect of E(2) on endothelial NO production, confluent bovine aortic endothelial cells (BAECs) were incubated in the absence or presence of E(2), and NO production was measured. Significant increases in NO levels were detected after only 5 min of E(2) exposure without a change in the protein levels of endothelial NO synthase (eNOS). This short-term effect of estrogen was significantly blunted by various ligands which decrease intracellular Ca(2+) concentration. Furthermore, plasma membrane-impermeable BSA-conjugated E(2) (E(2)BSA) stimulated endothelial NO release, indicating that in the current system the site of action of E(2) is on the plasma membrane rather than the classical nuclear receptor. The partial antagonist tamoxifen did not block E(2)-induced NO production; however, a pure estrogen receptor alpha (ERalpha) antagonist ICI 182,780 completely inhibited E(2)-stimulated NO release. The binding of E(2) to the membrane was confirmed using FITC-labeled E(2)BSA (E(2)BSA-FITC). Western blot analysis showed that plasmalemmal caveolae possess ERalpha in addition to well-known caveolae-associated proteins eNOS and caveolin. This study demonstrates that the nongenomic and short-term effect of E(2) on endothelial NO release is Ca(2+)-dependent and occurs via ERalpha localized in plasmalemmal caveolae.
Collapse
Affiliation(s)
- H P Kim
- Department of Biomedical Sciences, National Institute of Health, 5 Nokbun-dong, Seoul, Eunpyung-ku, [122-701], Korea
| | | | | | | | | | | |
Collapse
|
309
|
Rupprecht R, Holsboer F. Neuroactive steroids: mechanisms of action and neuropsychopharmacological perspectives. Trends Neurosci 1999; 22:410-6. [PMID: 10441302 DOI: 10.1016/s0166-2236(99)01399-5] [Citation(s) in RCA: 455] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Steroids influence neuronal function by binding to intracellular receptors that can act as transcription factors and regulate gene expression. In addition, some so-called 'neuroactive steroids' are potent modulators of an array of ligand-gated ion channels and of distinct G-protein coupled receptors via nongenomic mechanisms, and they can influence sleep and memory. This article describes how these neuroactive steroids modulate neurotransmitter receptors and addresses the neuropsychopharmacological potential that arises from the intracellular crosstalk between genomic and nongenomic steroid effects. Neuroactive steroids could also have a role in the response to stress and the treatment of psychiatric disorders, such as depression, and, as they affect a broad spectrum of behavioral functions through their unique molecular properties, they could constitute a yet unexploited class of drugs.
Collapse
Affiliation(s)
- R Rupprecht
- Dept of Psychiatry, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | | |
Collapse
|
310
|
Alarid ET, Bakopoulos N, Solodin N. Proteasome-mediated proteolysis of estrogen receptor: a novel component in autologous down-regulation. Mol Endocrinol 1999; 13:1522-34. [PMID: 10478843 DOI: 10.1210/mend.13.9.0337] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Regulation of estrogen receptor (ER) concentration is a key component in limiting estrogen responsiveness in target cells. Yet the mechanisms governing ER concentration in the lactotrope cells of the anterior pituitary, a major site of estrogen action, are undetermined. In this study, we used a lactotrope cell line, PR1, to explore regulation of ER protein by estrogen. Estrogen treatment resulted in an approximate 60% decrease in ER steady state protein levels. Suprisingly, the decline in ER protein was apparent within 1 h of estrogen treatment and occurred in the absence of protein synthesis and transcription. Direct regulation of ER protein was further confirmed by pulse chase analysis, which showed that ER protein half-life was shortened from greater than 3 h to 1 h in the presence of estrogen. The estrogen-induced degradation of ER protein could be prevented by pretreatment with peptide aldehyde inhibitors of proteasome protease whereas inhibitors of calpain and lysosomal proteases were ineffective. Inhibition of proteasome activity maintained ER protein at a level equivalent to control cells not stimulated with estrogen but increased estrogen-binding activity by 1.75-fold. Proteolytic regulation of ER by the proteasome is not limited to pituitary lactotrope cells but is also operational in MCF-7 breast cancer cells, suggesting that this may be a common regulatory pathway used by estrogen. These studies describe a nongenomic action of estrogen that involves nuclear ER: rapid proteolysis of ER protein via a proteasome-mediated pathway.
Collapse
Affiliation(s)
- E T Alarid
- The Department of Physiology, University of Wisconsin-Madison, 53706, USA.
| | | | | |
Collapse
|
311
|
Norfleet AM, Thomas ML, Gametchu B, Watson CS. Estrogen receptor-alpha detected on the plasma membrane of aldehyde-fixed GH3/B6/F10 rat pituitary tumor cells by enzyme-linked immunocytochemistry. Endocrinology 1999; 140:3805-14. [PMID: 10433242 DOI: 10.1210/endo.140.8.6936] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A population of estrogen receptor-alpha (ER alpha) proteins, located at the plasma membrane, is postulated to mediate the rapid, nongenomic responses of GH3/B6/F10 pituitary cells to estrogen. To demonstrate the presence of ER alpha at the plasma membrane and to distinguish this receptor population from that in the nucleus, GH3/B6/F10 cells were first prepared in 2% paraformaldehyde/0.1% glutaraldehyde in PBS (P/G) without detergent, then exposed to one of several antibodies (Abs) raised against nuclear ER alpha. Ab binding was visualized as a fluorescent/chromagenic reaction product catalyzed by avidin-biotin-complexed alkaline phosphatase. With P/G fixation, Abs could only access antigens at the cell surface, as evidenced by the inability of 70K mol wt dextrans to permeate cells and the absence of intracellular staining by Abs to cytoplasmic or nuclear antigens. ER alpha Abs generated membrane, but not nuclear, staining in P/G-fixed cells; nuclear receptor labeling could only be detected in detergent-treated cells. Specificity of staining for ER alpha was confirmed by three approaches: first, treatment with an antisense oligodeoxynucleotide to nuclear ER alpha mRNA reduced immunolabeling of both membrane and nuclear ER alpha; second, labeling by two Abs raised against different ER alpha oligopeptides was neutralized by competing peptide; third, six Abs (ER21, H226, R4, H222, MC20, and C542) that recognize unique epitopes on rodent ER alpha produced immunolabeling, but neither primate-specific ER alpha Ab nor Ab to ER beta caused staining. In addition to demonstrating the plasma membrane ER alpha in GH3/B6/F10 cells, this method should be applicable to other cell types that exhibit nongenomic responses to estrogen or other steroid hormones.
Collapse
Affiliation(s)
- A M Norfleet
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston 77555-0645, USA
| | | | | | | |
Collapse
|
312
|
Affiliation(s)
- M E Mendelsohn
- Molecular Cardiology Research Institute and the Department of Medicine, New England Medical Center and Tufts University School of Medicine, Boston, MA 02111, USA.
| | | |
Collapse
|
313
|
Paria BC, Tan J, Lubahn DB, Dey SK, Das SK. Uterine decidual response occurs in estrogen receptor-alpha-deficient mice. Endocrinology 1999; 140:2704-10. [PMID: 10342861 PMCID: PMC4273314 DOI: 10.1210/endo.140.6.6825] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Embryo-uterine interactions leading to the attachment reaction is followed by stromal cell proliferation and differentiation into decidual cells (decidualization) at the sites of blastocyst apposition. In rodents, decidualization is also induced by application of an artificial stimulus (intraluminal oil infusion) in a pseudopregnant uterus, or to one that has been appropriately prepared by exogenous progesterone (P4) and estrogen. The process of decidualization is under the control of these steroids in the presence of blastocysts or deciduogenic stimuli. Although it is well known that estrogen is required for the induction of progesterone receptors in the uterus, the functional importance of estrogen in the process of decidualization is poorly understood. To better understand the role of estrogenic actions in decidualization, we used wild-type and estrogen receptor-alpha knock-out (ERKO) mice for induction of decidualization employing a defined steroid hormonal treatment schedule. Our results demonstrate that P4 alone induces decidualization in ovariectomized wild-type or ERKO mice in response to intraluminal oil infusion in the absence of estrogen. A combined treatment of either estradiol-17beta (E2) or its catecholmetabolite 4-hydroxyestradiol-17beta(4-OH-E2) with P4 does not potentiate the decidual response produced by P4 treatment alone in either ovariectomized wild-type or ERKO mice. The induction of decidual response was associated with up-regulation of decidual cell marker genes, such as progesterone receptor, metallothionein-1, and cyclooxygenase-2. The results suggest that the stromal cell sensitivity to decidualization is critically dependent on P4-regulated events, and estrogenic induction of progesterone receptor via classical nuclear ER-alpha is not critical for this process.
Collapse
Affiliation(s)
- B C Paria
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City 66160-7338, USA
| | | | | | | | | |
Collapse
|
314
|
Abstract
All scientific investigations begin with distinct objectives: first is the hypothesis upon which studies are undertaken to disprove, and second is the overall aim of obtaining further information, from which future and more precise hypotheses may be drawn. Studies focusing on the generation and use of gene-targeted animal models also apply these goals and may be loosely categorized into sequential phases that become apparent as the use of the model progresses. Initial studies of knockout models often focus on the plausibility of the model based on prior knowledge and whether the generation of an animal lacking the particular gene will prove lethal or not. Upon the successful generation of a knockout, confirmatory studies are undertaken to corroborate previously established hypotheses of the function of the disrupted gene product. As these studies continue, observations of unpredicted phenotypes or, more likely, the lack of a phenotype that was expected based on models put forth from past investigations are noted. Often the surprising phenotype is due to the loss of a gene product that is downstream from the functions of the disrupted gene, whereas the lack of an expected phenotype may be due to compensatory roles filled by alternate mechanisms. As the descriptive studies of the knockout continue, use of the model is often shifted to the role as a unique research reagent, to be used in studies that 1) were not previously possible in a wild-type model; 2) aimed at finding related proteins or pathways whose existence or functions were previously masked; or 3) the subsequent effects of the gene disruption on related physiological and biochemical systems. The alpha ERKO mice continue to satisfy the confirmatory role of a knockout quite well. As summarized in Table 4, the phenotypes observed in the alpha ERKO due to estrogen insensitivity have definitively illustrated several roles that were previously believed to be dependent on functional ER alpha, including 1) the proliferative and differentiative actions critical to the function of the adult female reproductive tract and mammary gland; 2) as an obligatory component in growth factor signaling in the uterus and mammary gland; 3) as the principal steroid involved in negative regulation of gonadotropin gene transcription and LH levels in the hypothalamic-pituitary axis; 4) as a positive regulator of PR expression in several tissues; 5) in the positive regulation of PRL synthesis and secretion from the pituitary; 6) as a promotional factor in oncogene-induced mammary neoplasia; and 7) as a crucial component in the differentiation and activation of several behaviors in both the female and male. The list of unpredictable phenotypes in the alpha ERKO must begin with the observation that generation of an animal lacking a functional ER alpha gene was successful and produced animals of both sexes that exhibit a life span comparable to wild-type. The successful generation of beta ERKO mice suggests that this receptor is also not essential to survival and was most likely not a compensatory factor in the survival of the alpha ERKO. In support of this is our recent successful generation of double knockout, or alpha beta ERKO mice of both sexes. The precise defects in certain components of male reproduction, including the production of abnormal sperm and the loss of intromission and ejaculatory responses that were observed in the alpha ERKO, were quite surprising. In turn, certain estrogen pathways in the alpha ERKO female appear intact or unaffected, such as the ability of the uterus to successfully exhibit a progesterone-induced decidualization response, and the possible maintenance of an LH surge system in the hypothalamus. [ABSTRACT TRUNCATED]
Collapse
Affiliation(s)
- J F Couse
- Receptor Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
315
|
|
316
|
Powell CE, Watson CS, Gametchu B. Immunoaffinity isolation of native membrane glucocorticoid receptor from S-49++ lymphoma cells: biochemical characterization and interaction with Hsp 70 and Hsp 90. Endocrine 1999; 10:271-80. [PMID: 10484291 DOI: 10.1007/bf02738626] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The membrane glucocorticoid receptor (mGR), previously correlated with glucocorticoid-induced lymphocytolytic competency, was purified under nondenaturing conditions from mGR-enriched mouse S-49 T lymphoma cells. Proteins were immunoaffinity batch adsorbed to BUGR-2 monoclonal antibody-coupled protein A Sepharose 4B beads, and elution by epitope competition was compared with standard denaturation procedures. Elution with BUGR-2 epitope peptides released multiple mGRs (42-150 kDa) and heat shock proteins 70 and 90, suggesting that mGR interacts with these protein chaperones under physiological conditions. The mGR-heat shock protein 90 interaction was inhibited by 1 microM geldanamycin. Several other mGR binding partners were captured and most were dissociated from mGR by 0.6 M salt. Peptide maps of purified mGR displayed immunoreactive bands unique to mGR. Scatchard analysis estimated a k(d) value of 239 nM and a Bmax of 384 fmol/mg protein for mGR, compared to a k(d) of 19.5 nM and a Bmax of 90.3 fmol/mg protein for the intracellular GR (iGR). The rank order of affinities for mGR were RU-486 > dexamethasone > triamcinolone acetonide = aldosterone. Other steroids had no significant binding affinity. These results show that epitope-purified mGR on the plasma membrane of mouse lymphoma cells is similar but not identical to iGR.
Collapse
Affiliation(s)
- C E Powell
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | |
Collapse
|
317
|
Christ M, Haseroth K, Falkenstein E, Wehling M. Nongenomic steroid actions: fact or fantasy? VITAMINS AND HORMONES 1999; 57:325-73. [PMID: 10232053 DOI: 10.1016/s0083-6729(08)60647-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the common theory of steroid action, steroids bind to intracellular receptors and modulate nuclear transcription after translocation of steroid--receptor complexes into the nucleus. Due to homologies of molecular structure, specific receptors for steroids, vitamin D3, and thyroid hormone are considered to represent a superfamily of steroid receptors. While genomic steroid effects being characterized by their delayed onset of action and their sensitivity to blockers of transcription and protein synthesis have been known for several decades, very rapid actions of steroids have been more widely recognized and characterized in detail only recently. Rapid effects of steroids, vitamin D3, and thyroid hormones on cellular signaling and function may be transmitted by specific membrane receptors. Although no receptor of this kind has been cloned up to now, binding sites in membranes have been characterized exposing binding features compatible with an involvement in rapid steroid signaling. Characteristics of putative membrane receptors were completely different from those of intracellular steroid receptors, which was further supported by the inability of classic steroid receptor antagonists to inhibit nongenomic steroid actions. Development of drugs that specifically affect nongenomic action alone or even both modes of actions may find applications in various areas such as the cardiovascular and central nervous systems and treatment of preterm labor, infertility, and electrolyte homeostasis. To acquaint the reader with major aspects of nongenomic steroid actions, these effects on cellular function will be summarized, potentially related binding sites in membranes discussed, and the physiological or pathophysiological relevance of nonclassic actions exemplified.
Collapse
Affiliation(s)
- M Christ
- Institute of Clinical Pharmacology, Faculty for Clinical Medicine at Mannheim, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
318
|
Luconi M, Muratori M, Forti G, Baldi E. Identification and characterization of a novel functional estrogen receptor on human sperm membrane that interferes with progesterone effects. J Clin Endocrinol Metab 1999; 84:1670-8. [PMID: 10323398 DOI: 10.1210/jcem.84.5.5670] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The presence of a novel functional estrogen receptor on the human sperm surface has been demonstrated by using different experimental approaches. Ligand blot analysis of sperm lysates, using peroxidase-conjugated estradiol as probe, identified a specific estradiol-binding protein of approximately 29-kDa apparent molecular mass. The same protein band was also revealed by using alphaH222 antibody, which is directed against the steroid binding domain of the genomic estrogen receptor. The biological effects of estrogen receptor were investigated by analyzing calcium fluxes, tyrosine phosphorylation, and acrosome reaction (AR) in response to 17beta-estradiol (17betaE2) and by measuring the steroid influence on calcium and AR in responses to progesterone (P), a well-known physiological stimulus for human spermatozoa. Our results demonstrate that 17betaE2 induces a rapid and sustained increase of intracellular calcium concentrations ([Ca2+]i). This effect is totally dependent on the presence of extracellular calcium, because it is completely abolished in a calcium-depleted medium. The dose-response curve for calcium increase to 17betaE2 is biphasic with a first component in the nanomolar range (effective concentration 50 = 0.60 +/- 0.12 nmol/L) and a second component in the micromolar range (EC50 = 3.80 +/- 0.26 micromol/L). 17BetaE2 stimulates tyrosine phosphorylation of several sperm proteins, including the 29-kDa protein band, and determines a reduction of calcium response to P, finally resulting in inhibition of P-stimulated sperm AR. Conversely, no direct effect of 17betaE2 is observed on AR. 17BetaE2 effects on calcium are clearly mediated by a membrane receptor, because they are reproduced by the membrane-impermeable conjugate of the hormone BSA-E2 and reduced by sperm preincubation with alphaH222 antibody. Taken together, our results clearly show the presence of a functional surface estrogen receptor, of 29 kDa, on human spermatozoa. This receptor may play a role in the modulation of nongenomic action of P in these cells during the process of fertilization.
Collapse
Affiliation(s)
- M Luconi
- Dipartimento di Fisiopatologia Clinica, Unita' di Andrologia, Università di Firenze, Florence, Italy.
| | | | | | | |
Collapse
|
319
|
|
320
|
Toran-Allerand CD, Singh M, Sétáló G. Novel mechanisms of estrogen action in the brain: new players in an old story. Front Neuroendocrinol 1999; 20:97-121. [PMID: 10328986 DOI: 10.1006/frne.1999.0177] [Citation(s) in RCA: 345] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Estrogen elicits a selective enhancement of the growth and differentiation of axons and dendrites (neurites) in the developing brain. Widespread colocalization of estrogen and neurotrophin receptors (trk) within estrogen and neurotrophin targets, including neurons of the cerebral cortex, sensory ganglia, and PC12 cells, has been shown to result in differential and reciprocal transcriptional regulation of these receptors by their ligands. In addition, estrogen and neurotrophin receptor coexpression leads to convergence or cross-coupling of their signaling pathways, particularly at the level of the mitogen-activated protein (MAP) kinase cascade. 17beta-Estradiol elicits rapid (within 5-15 min) and sustained (at least 2 h) tyrosine phosphorylation and activation of the MAP kinases, extracellular-signal regulated kinase (ERK)1, and ERK2, which is successfully inhibited by the MAP kinase/ERK kinase 1 inhibitor PD98059, but not by the estrogen receptor (ER) antagonist ICI 182,780 and also does not appear to result from estradiol-induced activation of trk. Furthermore, the ability of estradiol to phosphorylate ERK persists even in ER-alpha knockout mice, implicating other estrogen receptors such as ER-beta in these actions of estradiol. The existence of an estrogen receptor-containing, multimeric complex consisting of hsp90, src, and B-Raf also suggests a direct link between the estrogen receptor and the MAP kinase signaling cascade. Collectively, these novel findings, coupled with our growing understanding of additional signaling substrates utilized by estrogen, provide alternative mechanisms for estrogen action in the developing brain which could explain not only some of the very rapid effects of estrogen, but also the ability of estrogen and neurotrophins to regulate the same broad array of cytoskeletal and growth-associated genes involved in neurite growth and differentiation. This review expands the usually restrictive view of estrogen action in the brain beyond the confines of sexual differentiation and reproductive neuroendocrine function. It considers the much broader question of estrogen as a neural growth factor with important influences on the development, survival, plasticity, regeneration, and aging of the mammalian brain and supports the view that the estrogen receptor is not only a ligand-induced transcriptional enhancer but also a mediator of rapid, nongenomic events.
Collapse
Affiliation(s)
- C D Toran-Allerand
- Department of Anatomy and Cell Biology, Center for Neurobiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | |
Collapse
|
321
|
Berger CE, Horrocks BR, Datta HK. Direct non-genomic effect of steroid hormones on superoxide anion generation in the bone resorbing osteoclasts. Mol Cell Endocrinol 1999; 149:53-9. [PMID: 10375017 DOI: 10.1016/s0303-7207(98)00261-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We have investigated the possible acute effect of steroid hormones, including 1alpha,25 dihydroxycholecalciferol (1alpha,25(OH)2D3) and estradiol, on the generation of superoxide anion (O2*-) in bone resorbing osteoclasts. Evidence is presented demonstrating acute non-genomic stimulatory action of 1alpha,25(OH)2D3 on the production of free radicals by rat osteoclasts cultured on calcified matrix. The increase in O2*- production was observed in the range of 6-10 s (n = 5) following exposure of enriched osteoclasts to 1alpha,25(OH)2D3 and was found to be transient with the peak response being in the range of 5-45 s (n = 5). The decline in the transient was much slower than the elevation, time for the decay being in the range 1-5 min (n = 5) and remained above the levels present prior to the addition. The exposure of the osteoclast to dexamethasone was found to have no effect on O2*- generation, whilst estradiol was found to be inhibitory. The mode of stimulation and the kinetics of the transients of O2*- in the bone resorbing osteoclasts produced by 1alpha,25(OH)2D3 were similar to that of parathyroid hormone (PTH) and pertussis. The exposure of the bone resorbing osteoclasts to cholera toxin was found to have no effect, suggesting that the stimulatory action is unlikely to be mediated via cAMP elevation. The importance of these observations is discussed in the context of calcium homeostasis and bone physiology.
Collapse
Affiliation(s)
- C E Berger
- Department of Clinical Biochemistry, The Medical School, University of Newcastle, Newcastle upon Tyne, UK
| | | | | |
Collapse
|
322
|
Christ M, Günther A, Heck M, Schmidt BM, Falkenstein E, Wehling M. Aldosterone, not estradiol, is the physiological agonist for rapid increases in cAMP in vascular smooth muscle cells. Circulation 1999; 99:1485-91. [PMID: 10086974 DOI: 10.1161/01.cir.99.11.1485] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Steroid-induced gene regulation in the endocrine tissues and vascular wall is achieved through the interaction of specific receptor proteins and promoters of target genes. In addition to these delayed steroid actions, rapid effects of steroids have been reported in various tissues that were clearly incompatible with the classic theory of genomic steroid action. METHODS AND RESULTS Because high doses of 17beta-estradiol have been shown to modulate intracellular cAMP levels in vascular smooth muscle cells, steroid-induced stimulation of adenylate cyclase stimulation and phosphorylation of cAMP response element binding protein was investigated in porcine coronary artery vascular smooth muscle cells. Aldosterone induces a approximately 1.5- to 2.5-fold increase in intracellular cAMP levels (EC50 approximately 0.01 to 0.1 nmol/L) within 1 minute, whereas 17beta-estradiol and hydrocortisone act only at supraphysiological concentrations (10 micromol/L). Aldosterone-induced changes in intracellular cAMP are calcium dependent; they are not blocked by inhibitors of mineralocorticoid receptors, transcription, or protein synthesis. In addition, aldosterone induces a time-dependent phosphorylation of cAMP response element binding protein with potential transcriptional importance. CONCLUSIONS A nongenomic modulation of vascular smooth muscle cells by aldosterone is consistent with the data that aldosterone, not estrogen, is the physiological stimulus for cAMP.
Collapse
Affiliation(s)
- M Christ
- Institute of Clinical Pharmacology, Klinikum Mannheim of the University of Heidelberg, Mannheim, Germany
| | | | | | | | | | | |
Collapse
|
323
|
Estrogen-induced activation of mitogen-activated protein kinase in cerebral cortical explants: convergence of estrogen and neurotrophin signaling pathways. J Neurosci 1999. [PMID: 9952396 DOI: 10.1523/jneurosci.19-04-01179.1999] [Citation(s) in RCA: 344] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have shown that estrogen elicits a selective enhancement of the growth and differentiation of axons and dendrites (neurites) in the developing CNS. We subsequently demonstrated widespread colocalization of estrogen and neurotrophin receptors (trk) within developing forebrain neurons and reciprocal transcriptional regulation of these receptors by their ligands. Using organotypic explants of the cerebral cortex, we tested the hypothesis that estrogen/neurotrophin receptor coexpression also may result in convergence or cross-coupling of their signaling pathways. Estradiol elicited rapid (within 5-15 min) tyrosine phosphorylation/activation of the mitogen-activated protein (MAP) kinases, ERK1 and ERK2, that persisted for at least 2 hr. This extracellular signal-regulated protein kinase (ERK) activation was inhibited successfully by the MEK1 inhibitor PD98059, but not by the estrogen receptor (ER) antagonist ICI 182,780, and did not appear to result from estradiol-induced activation of trk. Furthermore, we also found that estradiol elicited an increase in B-Raf kinase activity. The latter and subsequent downstream events leading to ERK activation may be a consequence of our documentation of a multimeric complex consisting of, at least, the ER, hsp90, and B-Raf. These novel findings provide an alternative mechanism for some of the estrogen actions in the developing CNS and could explain not only some of the very rapid effects of estrogen but also the ability of estrogen and neurotrophins to regulate the same broad array of cytoskeletal and growth-associated genes involved in neurite growth and differentiation.
Collapse
|
324
|
Russo J, Ao X, Grill C, Russo IH. Pattern of distribution of cells positive for estrogen receptor alpha and progesterone receptor in relation to proliferating cells in the mammary gland. Breast Cancer Res Treat 1999; 53:217-27. [PMID: 10369068 DOI: 10.1023/a:1006186719322] [Citation(s) in RCA: 253] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Since cell proliferation is indispensable for the growth and development of the breast, and estrogens are considered to play a major role in promoting cell proliferation, while progesterone influences its differentiation, the present work was designed with the purpose of verifying the relationship between cells containing steroid hormone receptors and proliferating cells in the normal human breast. Twelve breast samples were analyzed for their content of lobules type 1 (Lob1), Lob2, Lob3, and Lob4, and the number of cells containing estrogen receptor alpha (ER-alpha), progesterone receptor (PgR), or expressing Ki67 antibody was determined by double immunocytochemical technique with specific antibodies. The highest percentage of ER-alpha, PgR, and Ki67 positive cells was found in Lob1, with a progressive reduction in the more differentiated Lob2 and Lob3. ER-alpha and PgR positive cells were found exclusively in the breast epithelium and were negative for Ki67, while cells positive for Ki67 did not express receptors. These findings were compared with the distribution of ER-alpha and PgR in the autoradiographs of mammary gland of young virgin rats inoculated with 3H-thymidine for determination of the DNA labeling index (DNA-LI). Both the DNA-LI and the percentage of ER-alpha and PgR positive cells were maximal in the epithelium of terminal end buds, and these values were reduced in alveolar buds and lobules. ER-alpha and PgR positive cells did not proliferate, and those cells that had incorporated 3H-thymidine were negative for both receptors. Our results led us to conclude that the content of ER-alpha and PgR in the normal mammary tissue varies with the degree of lobular development, in parallel with cell proliferation. However, the expression of receptors occurs in cells other than the proliferating cells, indicating that they represent at least two separate cell populations. These findings open new avenues towards the understanding of the mechanisms through which estrogens and progesterone affect the proliferative activity of breast epithelial cells, and their role in the initiation of the cascade of events that leads a normal cell to cancer.
Collapse
Affiliation(s)
- J Russo
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|
325
|
Gu Q, Korach KS, Moss RL. Rapid action of 17beta-estradiol on kainate-induced currents in hippocampal neurons lacking intracellular estrogen receptors. Endocrinology 1999; 140:660-6. [PMID: 9927291 DOI: 10.1210/endo.140.2.6500] [Citation(s) in RCA: 141] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
17Beta-estradiol can potentiate kainate-induced currents in isolated hippocampal CA1 neurons. The action of estrogen was rapid in onset, steroid and stereospecific, and reversible. The potentiation could be mimicked by 8-bromo-cAMP, an activator of protein kinase A. As the hippocampus expresses both isoforms of the intracellular estrogen receptor (ER alpha and ER beta), the role of ERs in the rapid action of 17beta-estradiol remains elusive. Here we report that the rapid action of 17beta-estradiol is independent from the classical ER activation in the modulation of membrane excitability. Under whole cell voltage clamp recording configuration, 17beta-estradiol-induced potentiation was observed in both wild-type and the ER alpha gene knockout mice. The perfusion or incubation of ICI 182,780, which blocks both ER alpha and ER beta, did not affect estrogen potentiation in either group. Further study showed that adenosine 3',5'-cyclic-monophosphothioate Rp-isomer, a specific inhibitor of protein kinase A, completely blocked the potentiation observed with the application of 17beta-estradiol in ER alpha gene knockout mice. Our results provide evidence that a distinct estrogen-binding site exists, which appears to be coupled to alpha-amino-3-hydroxyl-5-methyl-4-isoxazole proprionic acid/kainate receptors by a cAMP-dependent phosphorylation process.
Collapse
Affiliation(s)
- Q Gu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas 75235-9049, USA
| | | | | |
Collapse
|
326
|
Razandi M, Pedram A, Greene GL, Levin ER. Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol 1999; 13:307-19. [PMID: 9973260 DOI: 10.1210/mend.13.2.0239] [Citation(s) in RCA: 273] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The existence of a putative membrane estrogen receptor (ER) has been supported by studies accomplished over the past 20 yr. However, the origin and functions of this receptor are not well defined. To study the membrane receptor, we transiently transfected cDNAs for ERalpha or ERbeta into Chinese hamster ovary (CHO) cells. Transfection of ERalpha resulted in a single transcript by Northern blot, specific binding of labeled 17beta-estradiol (E2), and expression of ER in both nuclear and membrane cell fractions. Competitive binding studies in both compartments revealed near identical dissociation constants (K(d)S) of 0.283 and 0.287 nM, respectively, but the membrane receptor number was only 3% as great as the nuclear receptor density. Transfection of ERbeta3 also yielded a single transcript and nuclear and membrane receptors with respective Kd values of 1.23 and 1.14 nM; the membrane receptor number was only 2% compared with expressed nuclear receptors. Estradiol binding to CHO-ERalpha or CHO-ERbeta activated Galphaq and G(alpha)s proteins in the membrane and rapidly stimulated corresponding inositol phosphate production and adenylate cyclase activity. Binding by 17-beta-E2 to either expressed receptor comparably enhanced the nuclear incorporation of thymidine, critically dependent upon the activation of the mitogen-activated protein kinase, ERK (extracellular regulated kinase). In contrast, c-Jun N-terminal kinase activity was stimulated by 17-beta-E2 in ERbeta-expressing CHO, but was inhibited in CHO-ERalpha cells. In summary, membrane and nuclear ER can be derived from a single transcript and have near-identical affinities for 17-beta-E2, but there are considerably more nuclear than membrane receptors. This is also the first report that cells can express a membrane ERbeta. Both membrane ERs activate G proteins, ERK, and cell proliferation, but there is novel differential regulation of c-Jun kinase activity by ERbeta and ERalpha.
Collapse
Affiliation(s)
- M Razandi
- Department of Medicine, University of California, Irvine 92717, USA
| | | | | | | |
Collapse
|
327
|
Abstract
Estrogens exert fast non-genomic actions in their target tissues which may involve the participation of receptors located at the cell membrane. Studies were performed to identify and characterize membrane-associated 17beta-estradiol binding proteins in rabbit uterus. Specific and saturable [3H]17beta-estradiol binding sites of high affinity (Kd = 0.36 nM) were detected in uterine microsomes at higher concentration than in cytosol (370 +/- 98 vs. 270 +/- 87 fmol/mg protein, respectively). Various other steroid hormones, the stereoisomer 17alpha-estradiol and the antiestrogen tamoxifen were significantly less effective than 17beta-estradiol to compete with the radioactive ligand for binding to the membranes. The microsome binding sites were trypsin-sensitive and could be extracted to a great extent (80-90%) with 0.4/0.6 M KCl. Assays of the marker enzyme glucose-6-P dehydrogenase excluded membrane contamination with cytosolic soluble components. Immunoblot analysis of particulate and soluble fractions using monoclonal antibodies against the transactivation, heat shock protein recognition, and steroid binding domains of the nuclear estrogen receptor (ER; 67 kDa), revealed lower concentrations of the ER in membranes and the presence of five additional immunoreactive proteins of 57, 50, 32, 28, and 11 kDa which were absent in cytosol. Moreover, the antibody against the steroid binding domain was as effective as an inhibitor for cytosolic and membrane specific radioligand binding. Extraction of microsomes with the nondenaturing detergent CHAPS allowed a 2-fold enrichment of ER-like binding proteins as shown by antibody labeling and [3H]17beta-estradiol binding analysis. The results of this work are consistent with the existence of novel 17beta-estradiol membrane binding proteins structurally related to the intracellular ER. Future studies should investigate whether any of these proteins are involved in the primary events (e.g. receptor function) mediating nongenomic estrogen effects.
Collapse
Affiliation(s)
- P Monje
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahia Blanca, Argentina
| | | |
Collapse
|
328
|
Benten WP, Lieberherr M, Giese G, Wrehlke C, Stamm O, Sekeris CE, Mossmann H, Wunderlich F. Functional testosterone receptors in plasma membranes of T cells. FASEB J 1999; 13:123-33. [PMID: 9872937 DOI: 10.1096/fasebj.13.1.123] [Citation(s) in RCA: 204] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
T cells are considered to be unresponsive to testosterone due to the absence of androgen receptors (AR). Here, we demonstrate the testosterone responsiveness of murine splenic T cells in vitro as well as the presence of unconventional cell surface receptors for testosterone and classical intracellular AR. Binding sites for testosterone on the surface of both CD4(+) and CD8(+) subsets of T cells are directly revealed with the impeded ligand testosterone-BSA-FITC by confocal laser scanning microscopy (CLSM) and flow cytometry, respectively. Binding of the plasma membrane impermeable testosterone-BSA conjugate induces a rapid rise (<5 s) in [Ca2+]i of Fura-2-loaded T cells. This rise reflects influx of extracellular Ca2+ through non-voltage-gated and Ni2+-blockable Ca2+ channels of the plasma membrane. The testosterone-BSA-induced Ca2+ import is not affected by cyproterone, a blocker of the AR. In addition, AR are not detectable on the surface of intact T cells when using anti-AR antibodies directed against the amino and carboxy terminus of the AR, although T cells contain AR, as revealed by reverse transcription-polymerase chain reactions and Western blotting. AR can be visualized with the anti-AR antibodies in the cytoplasm of permeabilized T cells by using CLSM, though AR are not detectable in cytosol fractions when using the charcoal binding assay with 3H-R1881 as ligand. Cytoplasmic AR do not translocate to the nucleus of T cells in the presence of testosterone, in contrast to cytoplasmic AR in human cancer LNCaP cells. These findings suggest that the classical AR present in splenic T cells are not active in the genomic pathway. By contrast, the cell surface receptors for testosterone are in a functionally active state, enabling T cells a nongenomic response to testosterone.
Collapse
Affiliation(s)
- W P Benten
- Division of Molecular Parasitology and Centre of Biological-Medical Research, Heinrich Heine University, 40225 Duesseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
329
|
Zheng J, Ramirez VD. Purification and identification of an estrogen binding protein from rat brain: oligomycin sensitivity-conferring protein (OSCP), a subunit of mitochondrial F0F1-ATP synthase/ATPase. J Steroid Biochem Mol Biol 1999; 68:65-75. [PMID: 10215039 DOI: 10.1016/s0960-0760(98)00161-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Early studies have suggested the presence in the central nervous system of possible estrogen binding sites/proteins other than classical nuclear estrogen receptors (nER). We report here the isolation and identification of a 23 kDa membrane protein from digitonin-solubilized rat brain mitochondrial fractions that binds 17beta-estradiol conjugated to bovine serum albumin at C-6 position (17beta-E-6-BSA), a ligand that also specifically binds nER. This protein was partially purified using affinity columns coupled with 17beta-E-6-BSA and was recognized by the iodinated 17beta-E-6-BSA (17beta-E-6-[125I]BSA) in a ligand blotting assay. The binding of 17beta-E-6-BSA to this protein was specific for the 17beta-estradiol portion of the conjugate, not BSA. Using N-terminal sequencing and immunoblotting, this 23 kDa protein was identified as the oligomycin-sensitivity conferring protein (OSCP). This protein is a subunit of the FOF1 (F-type) mitochondrial ATP synthase/ATPase required for the coupling of a proton gradient across the F0 sector of the enzyme in the mitochondrial membrane to ATP synthesis in the F1 sector of the enzyme. Studies using recombinant bovine OSCP (rbOSCP) in ligand blotting revealed that rbOSCP bound 17beta-E-6-[125I]BSA with the same specificity as the purified 23 kDa protein. Further, in a ligand binding assay, 17beta-E-6-[125I]BSA also bound rbOSCP and it was displaced by both 17beta-E-6-BSA and 17alpha-E-6-BSA as well as partially by 17beta-estradiol and diethylstilbestrol (DES), but not by BSA. This finding opens up the possibility that estradiol, and probably other compounds with similar structures, in addition to their classical genomic mechanism, may interact with ATP synthase/ATPase by binding to OSCP, and thereby modulating cellular energy metabolism. Current experiments are addressing such an issue.
Collapse
Affiliation(s)
- J Zheng
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana 61801, USA
| | | |
Collapse
|
330
|
Judy BM, Welshons WV. Cellular Localization of Receptors Mediating the Actions of Steroid Hormones. Compr Physiol 1998. [DOI: 10.1002/cphy.cp070117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
331
|
Jovanović S, Jovanović A, Shen WK, Terzic A. Protective action of 17beta-estradiol in cardiac cells: implications for hyperkalemic cardioplegia. Ann Thorac Surg 1998; 66:1658-61. [PMID: 9875767 DOI: 10.1016/s0003-4975(98)00893-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Hyperkalemic cardioplegic solutions effectively arrest the heart, but may also induce intracellular Ca2+ loading and cellular hypercontracture, which could contribute to ventricular dysfunction associated with global surgical ischemia. Recently, it has been proposed that 17beta-estradiol may possess protective properties in the ischemic myocardium. The purpose of the present study was to examine the action of 17beta-estradiol on cardiac cells exposed to hyperkalemic stress. METHODS Single ventricular cardiomyocytes, a preparation devoid of vascular and neuronal elements, were isolated from guinea pig hearts, loaded with a Ca2+-sensitive fluorescent probe, and imaged by digital epifluorescent microscopy. The emitted fluorescence of the probe, a measure of intracellular Ca2+ concentration, and cell length were simultaneously recorded during hyperkalemic challenge, in the absence or presence of 17beta-estradiol. RESULTS In control cardiomyocytes, the cytosolic concentration of Ca2+ was 138+/-11 nmol/L and cell length 93+/-11 microm. Exposure to high K+ (+16 mmol/L KCl) significantly increased cytosolic Ca2+ to 2,191+/-87 nmol/L (p < 0.001), and produced cell shortening (length at 39+/-5 microm; p < 0.001). 17beta-Estradiol (10 micromol/L) acutely prevented high K+ to induce either intracellular Ca2+ loading (144+/-13 nmol/L, p < 0.001) or hypercontracture (91+/-10 microm, p < 0.001). Tamoxifen (10 micromol/L), an antiestrogen, abolished the protective effect of 17beta-estradiol. CONCLUSIONS We conclude that 17beta-estradiol prevents hyperkalemia-induced Ca2+ loading and hypercontracture through a direct and tamoxifen-sensitive action in cardiomyocytes. This study raises the possibility that 17beta-estradiol should be considered as a cardioprotective adjunct toward a safer hyperkalemic cardioplegia.
Collapse
Affiliation(s)
- S Jovanović
- Department of Medicine, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
332
|
Nadal A, Rovira JM, Laribi O, Leon-quinto T, Andreu E, Ripoll C, Soria B. Rapid insulinotropic effect of 17beta-estradiol via a plasma membrane receptor. FASEB J 1998; 12:1341-8. [PMID: 9761777 DOI: 10.1096/fasebj.12.13.1341] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Impaired insulin secretion is a hallmark in both type I and type II diabetic individuals. Whereas type I (insulin-dependent diabetes mellitus) implies ss-cell destruction, type II (non-insulin dependent diabetes mellitus), responsible for 75% of diabetic syndromes, involves diminished glucose-dependent secretion of insulin from pancreatic beta-cells. Although a clear demonstration of a direct effect of 17beta-estradiol on the pancreatic ss-cell is lacking, an in vivo insulinotropic effect has been suggested. In this report we describe the effects of 17beta-estradiol in mouse pancreatic ss-cells. 17beta-Estradiol, at physiological concentrations, closes K(ATP) channels, which are also targets for antidiabetic sulfonylureas, in a rapid and reversible manner. Furthermore, in synergy with glucose, 17beta-estradiol depolarizes the plasma membrane, eliciting electrical activity and intracellular calcium signals, which in turn enhance insulin secretion. These effects occur through a receptor located at the plasma membrane, distinct from the classic cytosolic estrogen receptor. Specific competitive binding and localization of 17beta-estradiol receptors at the plasma membrane was demonstrated using confocal reflective microscopy and immunocytochemistry. Gaining deeper knowledge of the effect induced by 17beta-estradiol may be important in order to better understand the hormonal regulation of insulin secretion and for the treatment of NIDDM. receptor.
Collapse
Affiliation(s)
- A Nadal
- Institute of Bioengineering and Department of Physiology, Miguel Hernández University, San Juan Campus, Alicante, Spain.
| | | | | | | | | | | | | |
Collapse
|
333
|
|
334
|
Sylvia VL, Hughes T, Dean DD, Boyan BD, Schwartz Z. 17beta-estradiol regulation of protein kinase C activity in chondrocytes is sex-dependent and involves nongenomic mechanisms. J Cell Physiol 1998; 176:435-44. [PMID: 9648931 DOI: 10.1002/(sici)1097-4652(199808)176:2<435::aid-jcp22>3.0.co;2-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
17Beta-estradiol (E2) regulates growth plate chondrocyte differentiation in both a sex- and cell maturation-dependent manner, and the sex-specific effects of E2 appear to be mediated in part by membrane events. In this study, we examined whether E2 regulates protein kinase C (PKC) in a cell-maturation and sex-specific manner and whether E2 uses a nongenomic mechanism in regulating this enzyme. In addition, we determined if PKC mediates the E2-dependent stimulation of alkaline phosphatase activity seen in chondrocytes. Confluent, fourth passage resting zone (RC) and growth zone (GC) chondrocytes from male and female rat costochondral cartilage were treated with 10(-10) to 10(-7) M E2. E2 caused a dose-dependent increase in PKC in RC and GC cells from female rats. Peak stimulation was at 90 min. Increased PKC was evident by 3 min in both RC and GC and was still evident in RC cells at 720 min, but in GC cells activity returned to baseline by 270 min. Actinomycin D had no effect at 9, 90, 270, or 720 min, but there was a small decrease in E2-stimulated PKC in RC treated with cycloheximide at 90 and 270 min and in GC treated for 90 min. E2 increased cytosolic and membrane PKC at 9 min and by 90 min promoted translocation of PKC activity from the cytosol to the membranous compartment of female RC cells. Antibodies specific for the alpha, beta, delta, epsilon, and zeta isoforms of PKC revealed that PKCalpha in female GC and RC cells is activated by E2. There was a small, but statistically significant, increase in PKC in male RC cells in response to E2, but it was not dose-dependent, and no effect of E2 was noted in male GC cells. 17Alpha-estradiol, an inactive isomer of E2, did not affect PKC specific activity in RC or GC cells from either female or male rats. Chelerythrine, a specific inhibitor of PKC, inhibited E2-dependent alkaline phosphatase activity, indicating that E2 mediates its rapid effects on alkaline phosphatase via PKC.
Collapse
Affiliation(s)
- V L Sylvia
- Department of Orthopaedics, The University of Texas Health Science Center at San Antonio, 78284-7774, USA
| | | | | | | | | |
Collapse
|
335
|
Nemere I, Farach-Carson MC. Membrane receptors for steroid hormones: a case for specific cell surface binding sites for vitamin D metabolites and estrogens. Biochem Biophys Res Commun 1998; 248:443-9. [PMID: 9703943 DOI: 10.1006/bbrc.1998.8492] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Steroid hormones, including vitamin D metabolites and estrogens, activate target cells through specific receptors that discriminate among ligands based upon recognition of distinct structural features. For both classes of ligands, cell surface and nuclear receptors co-exist in many target cells. Upon ligand binding, these receptors generate both rapid and long lasting responses. While the structure of the nuclear receptors and their function as transcriptional activators of specific target genes is generally understood, the identity of the membrane receptors remains elusive. Using pharmacological, functional and biochemical approaches, new insights are being gained into nature of the cell surface receptors for both vitamin D metabolites and estrogens.
Collapse
Affiliation(s)
- I Nemere
- Department of Nutrition and Food Sciences, Utah State University, Logan 84322, USA
| | | |
Collapse
|
336
|
Mikkola T, Viinikka L, Ylikorkala O. Estrogen and postmenopausal estrogen/progestin therapy: effect on endothelium-dependent prostacyclin, nitric oxide and endothelin-1 production. Eur J Obstet Gynecol Reprod Biol 1998; 79:75-82. [PMID: 9643408 DOI: 10.1016/s0301-2115(98)00050-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is well documented that postmenopausal estrogen/progestin therapy (HRT) protects women against cardiovascular disorders. However, the mechanism(s) by which this protection is mediated remains largely unresolved, because beneficial effects of estrogen on the blood lipid profile account for only 20-30% of the overall protection. Growing evidence suggests that estrogen has direct effects on the blood vessel wall indicating that vascular endothelium may play a key role in mediating these effects by producing vasoactive factors, such as prostacyclin (PGI2), nitric oxide (NO) and endothelin-1 (ET-1). In vitro estrogen stimulates endothelial PGI2 and NO production, whereas ET-1 production is not affected. Moreover, in vivo studies indicate that estrogen and HRT increase PGI2 and NO production, whereas ET-1 production decreases. These effects are evidently mediated through estrogen receptors in endothelial cells. Thus, estrogen and HRT lead to the dominance of vasodilatory and antiaggregatory agents released by the endothelial cells. This may be an important new mechanism in the cardiovascular protection mediated by estrogen and HRT.
Collapse
Affiliation(s)
- T Mikkola
- Department of Obstetrics and Gynaecology, Helsinki University Central Hospital, Finland
| | | | | |
Collapse
|
337
|
Durkee TJ, Mueller M, Zinaman M. Identification of estrogen receptor protein and messenger ribonucleic acid in human spermatozoa. Am J Obstet Gynecol 1998; 178:1288-97. [PMID: 9662314 DOI: 10.1016/s0002-9378(98)70335-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The processes leading to fertilization involve a series of sequential events including the deposition and transport of sperm in the female genital tract. It is becoming evident that spermatozoa in which a hyperactive state has been induced are more effective in both reaching and penetrating the oocyte. Many of the changes that spermatozoa undergo are the result of their ability to respond to the milieu of the female genital tract. In the presence of estrogen sperm have been shown to have increased their metabolic activity and flagellar activity and to have an increased ability to penetrate oocytes. Most important, these observed changes in sperm physiology occur quickly, suggesting a novel second-messenger system coupled to the estrogen receptor. Established effects of steroid hormones involve mediation of the signal through genomic expression. However, because it has not been definitively demonstrated whether the human sperm express the estrogen receptor, the mechanism by which estrogen exert its effect remains to be elucidated. STUDY DESIGN The presence of estrogen receptors on human spermatozoa was investigated. Immunohistochemistry performed on human spermatozoa indicates that the estrogen receptors are located on the tailpiece. In addition, protein from human spermatozoa was isolated and subjected to Western blot analysis. RESULTS Results indicate a single band of approximately 65 kd, similar to that of the native human estrogen receptor. Ribonucleic acid obtained from the human spermatozoa was reverse transcribed into deoxyribonucleic acid. With use of selected primers, this deoxyribonucleic acid was amplified by polymerase chain reaction. Resolution and examination of the expansion products demonstrated a single band of deoxyribonucleic acid of 450 bp, identical to that expected from the selected primers. The specificity of this reverse transcriptase-polymerase chain reaction amplified deoxyribonucleic acid sequence was verified by Southern blotting. CONCLUSION For the first time we provide evidence as to the expression of estrogen receptor by human spermatozoa.
Collapse
Affiliation(s)
- T J Durkee
- Department of Obstetrics and Gynecology, Rockford Clinic, Illinois, USA
| | | | | |
Collapse
|
338
|
Abstract
Two osteoblastic cell populations, calvarial and marrow stromal cells, were exposed to estrogen derivatives in vitro. The hormonal effect was monitored by following intracellular Ca+2 levels [Ca+2]i and gap-junction communication. We measured fast changes in intracellular Ca+2 levels in response, of these cells, to the steroid hormones. The changes were dose dependent revealing maximal activity at 100 pM by 17-beta-Estradiol and 1 nM by estradiol-CMO. Additionally, the effect of estrogen, on functional coupling of the cells, was measured using fluorescence dye migration and counting the number of neighboring cells coupled by gap junctions. An uncoupling effect was demonstrated in response of these cells to estrogen treatment. The quick stereospecific effect was achieved in the presence of 17-beta-estradiol but not in the presence of 17-alpha-estradiol. These results suggest the involvement of plasma membrane receptors in addition to the already known nuclear receptors in transducing the hormone effects in the osteoblastic cells.
Collapse
Affiliation(s)
- R Massas
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | | | | |
Collapse
|
339
|
Ruehlmann DO, Steinert JR, Valverde MA, Jacob R, Mann GE. Environmental estrogenic pollutants induce acute vascular relaxation by inhibiting L-type Ca2+ channels in smooth muscle cells. FASEB J 1998; 12:613-9. [PMID: 9576488 DOI: 10.1096/fasebj.12.7.613] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There is an ongoing scientific debate concerning the potential threat of environmental estrogenic pollutants to animal and human health (1-5). Pollutants including the detergents 4-octylphenol and p-nonylphenol and chlorinated insecticides have recently been reported to modulate sexual differentiation by interacting with nuclear steroid receptors (6-8). So far, the focus has been on reproductive organs, but sex steroids have far more widespread actions. The lower incidence of cardiovascular disease in women has been attributed to estrogens (9-14), yet no information is available on the vascular actions of environmental estrogenic pollutants. In the present study we have investigated the effects of acute exposure to 17beta-estradiol, the antiestrogen ICI 182,780, and estrogenic pollutants on coronary vascular tone as well as on intracellular Ca2+ levels ([Ca2+]i) and Ca2+ and K+ channel activity in vascular smooth muscle cells. We report here that 4-octylphenol, p-nonylphenol, o.p'-DDT, and the antiestrogen ICI 182,780 inhibit L-type Ca2+ channels in vascular smooth muscle cells and evoke a rapid and endothelium-independent relaxation of the coronary vasculature similar to that induced by 17beta-estradiol. Thus, inhibition of Ca2+ influx via L-type Ca2+ channels in vascular smooth muscle cells may explain the acute, nongenomic vasodilator actions of environmental estrogenic pollutants.
Collapse
Affiliation(s)
- D O Ruehlmann
- Vascular Biology Research Centre, Biomedical Sciences Division, King's College London, UK
| | | | | | | | | |
Collapse
|
340
|
Abstract
Steroid hormones influence the electrical activity of many neurons and effectors by regulating the transcription of their ion channels and neurotransmitter receptors, or by modulating the activity of their channels and receptors through second messenger-coupled membrane receptors, or both. In this article, four cell types with known functions and distinct electrical activities are focused on to illustrate how different steroids act synergistically with, or in opposition to, each other to modulate specific electrical phenomena such as spontaneous regular firing (GH3 cells, a pituitary cell line), action potential duration (electric organ cells), and intrinsic excitability and sensitivity to neurotransmitters (GnRH and opioidergic neurons).These examples illustrate how steroids might influence electrical activity in neurons involved in more complex central circuits.
Collapse
Affiliation(s)
- H H Zakon
- Dept of Zoology, Patterson Laboratory, University of Texas, Austin 78712, USA
| |
Collapse
|
341
|
Affiliation(s)
- U Gehring
- Institut für Biologische Chemie, Universität Heidelberg, Germany
| |
Collapse
|
342
|
Marino M, Pallottini V, Trentalance A. Estrogens cause rapid activation of IP3-PKC-alpha signal transduction pathway in HEPG2 cells. Biochem Biophys Res Commun 1998; 245:254-8. [PMID: 9535818 DOI: 10.1006/bbrc.1998.8413] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanisms through which steroids affect target cells are not fully understood. In addition to the classic model, there is now increasing evidence that steroids can exert rapid actions. It must still be elucidated if rapid and slow estrogen actions produce co-operative and/or integrative functions. The effects of estrogen on inositol trisphosphate (IP3) production and PKC-alpha levels on membrane in the HEPG2 cell line have been investigated. Results show that estrogen addition to HEPG2 cells causes a rapid increase of IP3 production. The effect was totally inhibited by pre-incubation with tyrosine-kinase inhibitor genisteine and with the anti-estrogen ICI 182,780. An increased PKC-alpha level on the membrane fraction was present 30 min after estrogen exposure. The strong signal could elicit a variety of cellular responses such as modulation of ion channel, stimulation of cell proliferation, and phosphorylation of cytosolic ER. The ability of estrogen to trigger IP3 production in human hepatoma cells is a novel aspect of estrogen action that requires the current model of hormone stimulation target cells to be revised.
Collapse
Affiliation(s)
- M Marino
- Department of Biologia, Università "Roma 3,", v.le Marconi, 446, Rome, 00146, Italy
| | | | | |
Collapse
|
343
|
Migliaccio A, Piccolo D, Castoria G, Di Domenico M, Bilancio A, Lombardi M, Gong W, Beato M, Auricchio F. Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J 1998; 17:2008-18. [PMID: 9524123 PMCID: PMC1170546 DOI: 10.1093/emboj/17.7.2008] [Citation(s) in RCA: 456] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanisms by which ovarian hormones stimulate growth of breast tumors are unclear. It has been reported previously that estrogens activate the signal-transducing Src/p21(ras)/Erk pathway in human breast cancer cells via an interaction of estrogen receptor (ER) with c-Src. We now show that progestins stimulate human breast cancer T47D cell proliferation and induce a similar rapid and transient activation of the pathway which, surprisingly, is blocked not only by anti-progestins but also by anti-estrogens. In Cos-7 cells transfected with the B isoform of progesterone receptor (PRB), progestin activation of the MAP kinase pathway depends on co-transfection of ER. A transcriptionally inactive PRB mutant also activates the signaling pathway, demonstrating that this activity is independent of transcriptional effects. PRB does not interact with c-Src but associates via the N-terminal 168 amino acids with ER. This association is required for the signaling pathway activation by progestins. We propose that ER transmits to the Src/p21(ras)/Erk pathway signals received from the agonist-activated PRB. These findings reveal a hitherto unrecognized cross-talk between ovarian hormones which could be crucial for their growth-promoting effects on cancer cells.
Collapse
Affiliation(s)
- A Migliaccio
- Istituto di Patologia Generale e Oncologia, Facoltà di Medicina e Chirurgia, II Università di Napoli, Largo S.Aniello a Caponapoli, 2, 80138 Napoli, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
344
|
Wagner CK, Silverman AJ, Morrell JI. Evidence for estrogen receptor in cell nuclei and axon terminals within the lateral habenula of the rat: Regulation during pregnancy. J Comp Neurol 1998. [DOI: 10.1002/(sici)1096-9861(19980316)392:3<330::aid-cne4>3.0.co;2-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
345
|
Benten WP, Lieberherr M, Giese G, Wunderlich F. Estradiol binding to cell surface raises cytosolic free calcium in T cells. FEBS Lett 1998; 422:349-53. [PMID: 9498814 DOI: 10.1016/s0014-5793(98)00039-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Fura-2 method is used to examine a possible action of 17beta-estradiol (E2) on [Ca2+]i of splenic T cells isolated from female C57BL/10 mice. E2 concentrations between 10 fM and 10 nM induce a rapid and dose-dependent increase in [Ca2+]i due to Ca2+ influx and release of Ca2+ from intracellular stores. Ca2+ influx is mediated by Ca2+ channels which are completely blockable by Ni2+ and partly by nifedipine. The antiestrogen tamoxifen does not inhibit the E2-induced rise in [Ca2+]i. Ca2+ influx and Ca2+ release from intracellular stores is also inducible by plasma membrane impermeable E2 conjugated to BSA. E2-BSA-FITC binds to the surface of T cells of both the CD4+ and CD8+ subset. Our data suggest a novel E2-signalling pathway in T cells which is not mediated through the classical nuclear estrogen receptor response but rather through putative plasma membrane receptors for E2.
Collapse
Affiliation(s)
- W P Benten
- Division of Molecular Parasitology and Center for Biological and Medical Research, Heinrich-Heine-University, Düsseldorf, Germany
| | | | | | | |
Collapse
|
346
|
Gu Q, Moss RL. Novel mechanism for non-genomic action of 17 beta-oestradiol on kainate-induced currents in isolated rat CA1 hippocampal neurones. J Physiol 1998; 506 ( Pt 3):745-54. [PMID: 9503335 PMCID: PMC2230751 DOI: 10.1111/j.1469-7793.1998.745bv.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
1. Using whole-cell voltage-clamp recordings of dissociated hippocampal CA1 neurones, we demonstrated that 17 beta-oestradiol rapidly potentiates kainate-induced currents when applied either to the outside or the inside of the neurone. However, when the steroid was conjugated to bovine serum albumin (E2-BSA), application to either the extracellular plasma membrane (E2-BSAout) or the cytosolic side of the cell (E2-BSAin) had no observable effect on kainate-induced currents. However, when applied stimultaneously to both sides of the plasma membrane, E2-BSA potentiated kainate-induced currents. 2. Application of E2-BSAout and GTP gamma S(in) potentiated kainate-induced currents. The potentiation of kainate-induced currents by 17 beta-oestradiol was occluded by cholera toxin pretreatment and appeared to be pertussis toxin insensitive. 3. E2-BSAin prolonged the effect of 8-bromoadenosine 3',5' cyclic monophosphate (8-bromo-cAMP) on kainate-induced currents. The recovery from the 8-bromo-cAMP response was found to be a function of the concentration of E2-BSAin. The application of ATP gamma S(in) occluded the effect of 17 beta-oestradiol. 4. These results suggest that the non-genomic action of 17 beta-oestradiol in the potentiation of kainate-induced currents is mediated via an action on Gs protein-coupled receptors. This operates in concert with an internal action of 17 beta-oestradiol on a cAMP-dependent phosphorylation.
Collapse
Affiliation(s)
- Q Gu
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas 75235, USA
| | | |
Collapse
|
347
|
Beyer C, Raab H. Nongenomic effects of oestrogen: embryonic mouse midbrain neurones respond with a rapid release of calcium from intracellular stores. Eur J Neurosci 1998; 10:255-62. [PMID: 9753134 DOI: 10.1046/j.1460-9568.1998.00045.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Evidence is emerging that oestrogen, besides acting via classical nuclear receptors, can rapidly influence the physiology of nerve cells through other mechanisms. Oestrogens have been shown to modulate the differentiation and function of embryonic midbrain dopaminergic neurones by stimulating neurite outgrowth, expression of tyrosine hydroxylase mRNA, dopamine uptake and release in spite of the fact that dopaminergic cells in the prenatal midbrain do not express the classical oestrogen receptor. This study therefore intended to unravel possible signal transduction pathways activated by oestrogen which might be associated with the above oestrogen effects. As a physiological second-messenger mechanism, we studied the influence of oestrogen on fluctuations of intracellular Ca2+ levels [Ca2+]i by microspectrofluorimetry of the Ca2+-sensitive indicator Fura-2, in primary cultures from embryonic mouse midbrains. 17Beta-estradiol (10 nM-1 pM) but not 17alpha-estradiol increased [Ca2+]i within 1-3 s in a dose-dependent way. Removal of extracellular Ca2+ abrogated K+-stimulated Ca2+ rise but did not affect 17beta-estradiol stimulation. Pretreatment of cells with thapsigargin (1 microM, 10 min), an inhibitor of Ca2+-pumping ATPases in the endoplasmic reticulum, abolished the 17beta-estradiol effect but not the K+-stimulated [Ca2+]i rise. Oestrogen effects on [Ca2+]i were completely mimicked by using a membrane-impermeant oestrogen-BSA construct. In order to identify oestrogen-sensitive cells, some cultures were subsequently immunostained for microtubule-associated protein II, tyrosine hydroxylase, or GABA. All oestrogen-sensitive cells were immunocytochemically characterized as neurones, and about half of these responsive neurones was found to be dopaminergic or GABAergic. These results demonstrate that 17beta-estradiol is capable of rapidly modulating physiological parameters of developing midbrain neurones by directly interacting with specific membrane binding sites coupled to a signal transduction mechanism that causes a calcium release from intracellular Ca2+ stores. It is suggested that oestrogen effects on differentiation and function of midbrain dopaminergic neurones are mediated by intracellular Ca2+ signalling.
Collapse
Affiliation(s)
- C Beyer
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany.
| | | |
Collapse
|
348
|
Hegele-Hartung C, Fritzemeier KH, Diel P. Effects of a pure antiestrogen and progesterone on estrogen-mediated alterations of blood flow and progesterone receptor expression in the aorta of ovariectomized rabbits. J Steroid Biochem Mol Biol 1997; 63:237-49. [PMID: 9459190 DOI: 10.1016/s0960-0760(97)00125-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is ample evidence from epidemiological studies that estrogen-replacement therapy protects postmenopausal women against cardiovascular disease. One explanation for this beneficial effect could be the improvement of blood flow under estrogen therapy. By using ultrasound and Doppler color flow mapping we demonstrated in the aorta of ovariectomized rabbits a significant dose-dependent increase in blood flow after treatment with 17beta-estradiol. An increase in blood flow was already observed within 1 h of estradiol treatment and lasted until the end of a 14-day treatment phase. Progesterone did not attenuate the effects of 17beta-estradiol on aortic blood flow. The pure estrogen receptor antagonist ZM 182780, however, dose-dependently reversed the effect of 17beta-estradiol on blood flow after the 14-day treatment phase, but was not able to antagonize the rapid 17beta-estradiol effect on blood flow after 1 h. After killing the animals mRNA and protein expression of the progesterone receptor (PR), a known estrogen-responsive gene in classic target organs, were examined. Analogous to the blood flow results the PR mRNA level increased dose-dependently after 17beta-estradiol treatment, whereas ZM 182780 was able to reverse this effect. Immunohistochemical localization of PR in the aortic wall revealed an increase in immunoreactivity in fibroblasts of the adventitia after 17beta-estradiol treatment. ZM 182780, and to a lesser degree progesterone, reversed the 17beta-estradiol-induced increase in PR immunoreactivity. PR immunoreactivity was further detected in endothelial and smooth muscle cells, but the various hormonal treatments had no discernible effect on the PR mRNA level in these cellular compartments. Our findings in the aorta of OVX rabbits suggest that (a) 17beta-estradiol exhibits a rapid effect on arterial tone, (b) the pure estrogen receptor antagonist ZM 182780 inhibits the 17beta-estradiol effect on blood flow and PR mRNA and (c) progesterone does not attenuate the beneficial effect of estrogens on arterial tone.
Collapse
|
349
|
Sackey FN, Watson CS, Gametchu B. Cell cycle regulation of membrane glucocorticoid receptor in CCRF-CEM human ALL cells: correlation to apoptosis. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:E571-83. [PMID: 9316448 DOI: 10.1152/ajpendo.1997.273.3.e571] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The human leukemic cell line (CCRF-CEM) and a subline enriched for the plasma membrane-resident glucocorticoid receptor (mGR) were studied for the influence of the cell cycle on the expression and function of mGR. Three synchronization procedures (double thymidine, colcemid, and combined thymidine-colcemid blocks) were used. Fluorescent microscopy and flow cytometry simultaneously assessed antibody-tagged mGR and DNA. In addition, mGR was quantitated and characterized by immunoprecipitation and immunoblotting. Apoptosis was assayed by DNA fragmentation (TUNEL assay) and by cell survival (trypan blue exclusion). All synchronization procedures demonstrated that progression from DNA replication (S) to the second growth phase and mitosis (G2/M) leads to cells having the highest levels of mGR expression and being highly glucocorticoid sensitive in the apoptosis assays: 32 and 80% sensitivity of wild type and mGR-enriched cells, respectively, compared with 12 and 30% sensitivity in asynchronous cells. Therefore, mGR expression appears to be cell cycle regulated, with its highest expression at late S-G2/M, when the cells are most sensitive to the lymphocytolytic effects of glucocorticoids.
Collapse
Affiliation(s)
- F N Sackey
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | |
Collapse
|
350
|
Morey AK, Pedram A, Razandi M, Prins BA, Hu RM, Biesiada E, Levin ER. Estrogen and progesterone inhibit vascular smooth muscle proliferation. Endocrinology 1997; 138:3330-9. [PMID: 9231785 DOI: 10.1210/endo.138.8.5354] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Estrogen (E) has been identified in epidemiologic and prospective studies to protect against the development of cardiovascular disease in women. It is unclear whether progesterone (P) is similarly beneficial. The mechanisms by which E or P might act are incompletely defined. One possibility is that sex steroids inhibit the proliferation of vascular smooth muscle, an early/important event in vascular pathology. We examined the ability of E and P to inhibit the growth of human umbilical vein smooth muscle cells (hUVSMC) in culture, when stimulated by serum or the mitogen, endothelin-1 (ET-1). Serum and ET-1 stimulated hVSMC cell numbers by approximately 110% and 43% respectively, compared with control, after 3 days in culture. This stimulation was maximally reversed 75% by E and 64% by P. No synergistic or additive effects of the two steroids were found. ET-1 and serum stimulated mitogen-activated protein kinase (MAP-K) and MAP-kinase kinase activities, and these were critical for mitogenesis. Mitogen-stimulated MAP-kinase kinase and MAP-K activities were significantly inhibited by either E or P. The steroids also inhibited mitogen-stimulated c-fos and c-myc, downstream targets for MAP-K action. Critical signaling and molecular events through which mitogens stimulate VSMC proliferation can be significantly inhibited by E or P, providing a potential cellular mechanism for their vascular protective actions.
Collapse
MESH Headings
- Blotting, Northern
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Calcium-Calmodulin-Dependent Protein Kinases/physiology
- Cell Division/drug effects
- Cell Division/physiology
- Cells, Cultured
- DNA/metabolism
- Endothelin-1/pharmacology
- Enzyme Activation
- Estrogens/pharmacology
- Female
- Gene Expression Regulation/drug effects
- Genes, fos/genetics
- Genes, myc/genetics
- Humans
- Mitogen-Activated Protein Kinase Kinases
- Mitogens/pharmacology
- Muscle, Smooth, Vascular/chemistry
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Pregnancy
- Progesterone/pharmacology
- Protein Kinases/metabolism
- Protein Kinases/physiology
- Proto-Oncogene Proteins c-fos/analysis
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- Proto-Oncogene Proteins c-myc/analysis
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Serum Albumin, Bovine/pharmacology
- Thymidine/metabolism
- Tritium
- Umbilical Veins/chemistry
- Umbilical Veins/cytology
- Umbilical Veins/drug effects
Collapse
Affiliation(s)
- A K Morey
- Division of Endocrinology, Long Beach Veteran Affairs Medical Center, California 90822, USA
| | | | | | | | | | | | | |
Collapse
|