51
|
Mermelstein PG. Membrane-localised oestrogen receptor alpha and beta influence neuronal activity through activation of metabotropic glutamate receptors. J Neuroendocrinol 2009; 21:257-62. [PMID: 19207809 PMCID: PMC2805164 DOI: 10.1111/j.1365-2826.2009.01838.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Until recently, the idea that oestradiol could affect cellular processes independent of nuclear oestrogen receptors (ERs) was controversial. This was despite the large number of carefully controlled studies performed both within and outside the nervous system demonstrating that oestrogens regulate various intracellular signalling pathways by acting at the membrane surface of cells and/or at biological rates incompatible with the time course of genomic-initiated events. At present, it is far less controversial that oestradiol acts at surface membrane receptors to regulate nervous system function. Recent studies have demonstrated that the classical intracellular ERs, ERalpha and ERbeta, are major players in mediating the actions of oestradiol on the membrane surface. This review focuses on one potential mechanism by which surface-localised ERalpha and ERbeta stimulate intracellular signalling events in cells of the nervous system. After oestradiol treatment, both ERalpha and ERbeta are capable of activating different classes of metabotropic glutamate receptors (mGluRs). Oestradiol activation of mGluRs is independent of glutamate, but requires expression of several different caveolin proteins to compartmentalise the different ERs with mGluRs into functional signalling microdomains. ER/mGluR signalling is a potential means by which oestrogens can both rapidly and for extended periods, influence a variety of intracellular signalling processes and behaviours.
Collapse
Affiliation(s)
- P G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
52
|
Abstract
17Beta-oestradiol is a pleiotropic hormone with organisational, activational and protective actions in both male and female mammals. It is responsible for numerous aspects of brain development, including sexual differentiation of the brain. The organisational effects of early oestrogen exposure are essential for long-lasting cognitive and behavioural functions. Oestradiol mediates its effects through several intracellular or membrane-associated receptor proteins. In the rodent cerebral cortex, oestrogen receptor (ER) expression, as demonstrated by hormone binding studies, is high early in postnatal life and declines precipitously as the animal approaches puberty. This decline is caused by a decreased expression of ERalpha mRNA. An understanding of the mechanisms involved in the regulation of ERalpha gene expression is critical for understanding the developmental as well as postpubertal expression of the ER. Despite recent data indicating the current hormone replacement therapies can be detrimental in older women, numerous animal studies have shown that the endogenous oestrogen, 17beta-oestradiol, is neuroprotective. Specifically, low levels of oestradiol protect the cortex from cell death caused by middle cerebral artery occlusion (MCAO). The attenuation of cell death by oestradiol in this model is mediated through an ERalpha-dependent mechanism. To this end, ERalpha expression is rapidly increased after MCAO, suggesting a return to the developmental programme of gene expression within neurones. One mechanism of suppressing gene expression is by the epigenetic modification of the promoter regions, which results in gene silencing. Of the epigenetic modifications utilised by cells, DNA methylation has been intensively studied as a mechanism by which genes are both permanently and reversibly silenced. Little is known about the mechanisms of ER gene regulation in the brain; however, in breast cancer cells, both ERalpha and ERbeta are down-regulated by promoter methylation, and subsequent binding of the methyl-CpG-binding protein, MeCP2. Data from our laboratory demonstrate that the promoters of the ERalpha gene are also methylated during development and after neuronal injury, suggesting a role of DNA methylation in regulating ER expression in the brain.
Collapse
Affiliation(s)
- M E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| | | |
Collapse
|
53
|
Lane PH. Estrogen receptors in the kidney: lessons from genetically altered mice. ACTA ACUST UNITED AC 2008; 5 Suppl A:S11-8. [PMID: 18395676 DOI: 10.1016/j.genm.2008.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Sex differences in human and animal models of kidney disease suggest that estrogen receptor (ER)-mediated events may modulate these processes. Genetically altered mice lacking one or both ERs provide a powerful tool to study these phenomena. OBJECTIVE This article examines sex differences in the kidney, particularly the role of ERs. METHODS To identify pertinent studies in genetically altered mice, a literature search was conducted on the MEDLINE database from January 1966 to July 2007, using the search terms estrogen receptor, kidney, and mice. Our group examined the effect of the ER-alpha knockout genotype on the kidney in streptozotocin-induced diabetes mellitus and compensatory kidney growth after uninephrectomy. RESULTS Female mice lacking ERa had reduced renal growth, including glomerular enlargement after 2 weeks of streptozotocin-induced diabetes mellitus and compensatory kidney growth 48 hours after uninephrectomy. CONCLUSION ER-mediated events influence kidney growth and disease in female mice.
Collapse
Affiliation(s)
- Pascale H Lane
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
54
|
Micevych PE, Mermelstein PG. Membrane estrogen receptors acting through metabotropic glutamate receptors: an emerging mechanism of estrogen action in brain. Mol Neurobiol 2008; 38:66-77. [PMID: 18670908 DOI: 10.1007/s12035-008-8034-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 06/18/2008] [Indexed: 12/27/2022]
Abstract
It has been over 60 years since the first studies have been published describing the effects of steroid hormones on brain function. For over 30 years, estrogen has been presumed to directly affect gene expression and protein synthesis through a specific receptor. More than 20 years ago, the first estrogen receptor was cloned and identified as a transcription factor. Yet, throughout their course of study, estrogens have also been observed to affect nervous system function via mechanisms independent of intracellular receptor regulation of gene expression. Up until recently, the membrane estrogen receptors responsible for these rapid actions have remained elusive. Recent studies have demonstrated that a large number of these rapid, membrane-initiated actions of estradiol are due to surface expression of classical estrogen receptors. This review focuses on the importance of membrane estrogen receptor interactions with metabotropic glutamate receptors for understanding rapid estradiol signaling mechanisms and downstream effectors, as well as their significance in a variety of physiological processes.
Collapse
Affiliation(s)
- Paul E Micevych
- Department of Neurobiology and Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
55
|
Wilson ME, Westberry JM, Prewitt AK. Dynamic regulation of estrogen receptor-alpha gene expression in the brain: a role for promoter methylation? Front Neuroendocrinol 2008; 29:375-85. [PMID: 18439661 PMCID: PMC2460564 DOI: 10.1016/j.yfrne.2008.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 02/29/2008] [Accepted: 03/04/2008] [Indexed: 12/31/2022]
Abstract
Estrogen has long been known to play an important role in coordinating the neuroendocrine events that control sexual development, sexual behavior and reproduction. Estrogen actions in other, non-reproductive areas of the brain have also been described. It is now known that estrogen can also influence learning, memory, and emotion and has neurotrophic and neuroprotective properties. The actions of estrogen are largely mediated through at least two intracellular estrogen receptors. Both estrogen receptor-alpha and estrogen receptor-beta are expressed in a wide variety of brain regions. Estrogen receptor-alpha (ERalpha), however, undergoes developmental and brain region-specific changes in expression. The precise molecular mechanisms that regulate its expression at the level of gene transcription are not well understood. Adding to the complexity of its regulation, the estrogen receptor gene contains multiple promoters that drive its expression. In the cortex in particular, the ERalpha mRNA expression is dynamically regulated during postnatal development and again following neuronal injury. Epigenetic modification of chromatin is increasingly being understood as a mechanism of neuronal gene regulation. This review examines the potential regulation of the ERalpha gene by such epigenetic mechanisms.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
56
|
Heinzlmann A, Köves K. The characteristic change in the distribution of S-100 immunoreactive folliculostellate cells in rat anterior pituitary upon long-term estrogen treatment is prevented by concomitant progesterone treatment. Endocrine 2008; 33:342-8. [PMID: 19082791 DOI: 10.1007/s12020-008-9096-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 06/26/2008] [Indexed: 10/21/2022]
Abstract
The presence of folliculostellate cells in the anterior pituitary was described 49 years ago. These cells give about 10% of the whole cell population and through their long processes they provide intrahypophyseal communication. The folliculostellate cells contain S-100 protein. Its immunostaining was used to identify these cells. It was previously found that the diethylstilbestrol treatment basically influences the morphology and function of the trophic hormone secreting as well as the folliculostellate cells. In the present experiment, we have studied whether a concomitant progesterone treatment can prevent or attenuate changes caused by diethylstilbestrol treatment in the distribution of folliculostellate, prolactin, and GH cells. Diethylstilbestrol alone induced the appearance of prolactinomas. Inside the prolactinomas, folliculostellate cells were scattered but outside the prolactinomas they formed a demarcation line. Inside the prolactinomas, there were only a few growth hormone immunoreactive cells but they surrounded the prolactinomas in a ring-like pattern. When diethylstilbestrol was implanted with progesterone, the changes being characteristic for diethylstilbestrol treatment, could not develop. Concomitant progesterone influence prevented morphological changes in the anterior pituitary. Progesterone alone had no effect. In accordance with the formation of prolactinomas, the plasma prolactin level was very high in diethylstilbestrol treated rats. Concomitant progesterone treatment prevented the effect of diethylstilbestrol. Progesterone alone did not influence the prolactin level. GH levels did not significantly differ in any groups.
Collapse
Affiliation(s)
- Andrea Heinzlmann
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Tuzoltó u. 58, Budapest 1094, Hungary
| | | |
Collapse
|
57
|
Ito K, Mochida K, Fujii K. Molecular cloning of two estrogen receptors expressed in the testis of the Japanese common goby, Acanthogobius flavimanus. Zoolog Sci 2008; 24:986-96. [PMID: 18088175 DOI: 10.2108/zsj.24.986] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2007] [Accepted: 06/17/2007] [Indexed: 11/17/2022]
Abstract
We isolated cDNA clones of two estrogen receptors (ER1 and ER2) from testis of the Japanese common goby (Acanthogobius flavimanus). The cDNAs of ER1 contained 3,796 nucleotides, including an open reading frame encoding 564 amino acids (Mr: 61.9 kDa); the cDNA for ER2 was 3,274 nucleotides long, with an open reading frame of 567 amino acids (Mr: 63.5 kDa). The deduced aminoacid sequences of ER1 and ER2 each had a characteristic ER structure consisting of five domains (A/B, DNA-binding, D, ligand-binding, and F) and were homologous to ERalpha and ERbeta of other vertebrates. We therefore named ER1 and ER2 as goby ERalpha (gERalpha) and goby ERbeta (gERbeta), respectively. The DNA- and ligand-binding domains in each gER showed high similarity to the corresponding domains of other vertebrates. Analysis of the distribution of gERalpha and gERbeta mRNAs in tissue by reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed that gERalpha was expressed at high levels in brain and testis, and gERbeta was expressed most strongly in testis. In situ hybridization showed that the mRNA of each gER was expressed mainly in the Sertoli cells of goby testis.
Collapse
Affiliation(s)
- Katsutoshi Ito
- National Research Institute of Fisheries and Environment of Inland Sea, Fisheries Research Agency, 2-17-5 Maruishi, Hatsukaichi, Hiroshima, Japan.
| | | | | |
Collapse
|
58
|
Sun H, Xu XL, Qu JH, Hong X, Wang YB, Xu LC, Wang XR. 4-Alkylphenols and related chemicals show similar effect on the function of human and rat estrogen receptor alpha in reporter gene assay. CHEMOSPHERE 2008; 71:582-588. [PMID: 18028983 DOI: 10.1016/j.chemosphere.2007.09.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 08/29/2007] [Accepted: 09/12/2007] [Indexed: 05/25/2023]
Abstract
Alkylphenols (APs) are widely used as important industrial materials and have attracted lots of attention because of their potential estrogenic activities. In this study, we developed human estrogen receptor alpha (hERalpha) and rat estrogen receptor alpha (rERalpha) mediated reporter gene assays and compared the estrogenic activity of APs and related chemicals based on the two ERalpha. Human breast cancer cell line MCF-7 was co-transfected with Gal4-fused hERalpha and corresponding reporter plasmid; African green monkey kidney cell line CV-1 was co-transfected with rERalpha and reporter gene. Both assays showed acceptable response to natural estrogen 17beta-estradiol (E2) with EC50 of 0.16 nM and 4.7 nM. Then the estrogenic activity of 4-APs, 4-phenylphenol and bisphenol-A were evaluated and compared with the effects of E2. The data suggested that test APs and related chemicals possessed weakly estrogenic activity and the activity of test APs increased with the increase of substituent size. This structure-activity relationship helped to infer the activity of chemicals with similar feature. Furthermore, test APs showed similar effect on the function of hERalpha and rERalpha. This consistency helped to extrapolate in vivo rodent data to human being when performing risk assessment of endocrine disruptors.
Collapse
Affiliation(s)
- Hong Sun
- Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | |
Collapse
|
59
|
Teng J, Wang ZY, Jarrard DF, Bjorling DE. Roles of estrogen receptor alpha and beta in modulating urothelial cell proliferation. Endocr Relat Cancer 2008; 15:351-64. [PMID: 18310301 PMCID: PMC3513362 DOI: 10.1677/erc.1.01255] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We reported previously that both subtypes of estrogen receptors, ERalpha and ERbeta, are expressed by human urothelial cells and mediate estrogen-induced cell proliferation in these cells. The aim of this study was to determine the extent to which each ER subtype contributes to urothelial cell proliferation and their possible involvement in the regulation of the cell cycle. We compared the expression of ERalpha and ERbeta mRNAs and protein quantitatively in primarily cultured human bladder urothelial cells obtained from six individuals with three immortalized urothelial (E6, E7, and UROtsa) and two bladder cancer cell lines (HTB-9 and T24). We found that all these cells express similar levels of ERbeta, but immortalized and cancer cells express much higher amounts of ERalpha than primary cells. Higher levels of ERalpha mRNA were also observed in the biopsies of bladder transitional cell carcinoma compared with sample from the same bladder unaffected by tumor. Using the ERalpha-selective agonist PPT, the ERbeta-selective agonist DPN, and specific small interfering RNA against ERalpha or ERbeta, we found that ERbeta predominantly mediates estrogen-induced G1/S transition and cell proliferation in the primary urothelial cells. By contrast, ERalpha predominantly mediates estrogen-induced G1/S transition and cell proliferation in bladder cancer cell lines. Furthermore, we found that 17beta-estradiol (E(2)) rapidly induces phosphorylation of extracellular signal-regulated kinases, but U0126, a mitogen-activated protein kinase kinase (MEK) inhibitor, does not affect E(2)-induced urothelial cell proliferation. E(2) up-regulated cyclin D1 and cyclin E expression in both the primary and bladder cancer cells, and the cancer cells have higher cyclin D1 and cyclin E expression during G0/G1 phases. Our data suggest that estrogen exerts its effects through different ER subtypes in urothelial cells. Increased expression of ERalpha may contribute to early induction of cyclin D1 and cyclin E during the cell cycle in bladder cancer cells.
Collapse
MESH Headings
- Blotting, Western
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/metabolism
- Carcinoma, Transitional Cell/secondary
- Cell Proliferation
- Cyclin D
- Cyclin E/metabolism
- Cyclins/metabolism
- DNA Primers/chemistry
- Estradiol/pharmacology
- Estrogen Receptor alpha/antagonists & inhibitors
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/antagonists & inhibitors
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Estrogens/pharmacology
- Flow Cytometry
- G1 Phase/drug effects
- G1 Phase/physiology
- Humans
- Immunoenzyme Techniques
- Ligands
- MAP Kinase Kinase Kinases/antagonists & inhibitors
- MAP Kinase Kinase Kinases/metabolism
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Nitriles/pharmacology
- Phenols
- Pyrazoles/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Response Elements
- Reverse Transcriptase Polymerase Chain Reaction
- S Phase/drug effects
- S Phase/physiology
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
- Urothelium/metabolism
Collapse
Affiliation(s)
- Jian Teng
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
60
|
Epigenetic regulation of the estrogen receptor alpha promoter in the cerebral cortex following ischemia in male and female rats. Neuroscience 2008; 152:982-9. [PMID: 18353557 DOI: 10.1016/j.neuroscience.2008.01.048] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/18/2008] [Accepted: 02/04/2008] [Indexed: 11/22/2022]
Abstract
Permanent middle cerebral artery occlusion (MCAO) causes neuronal cell death in the striatum and cortex. In rodents, estradiol treatment protects the cortex from cell death in an estrogen receptor alpha (ERalpha) dependent manner. ERalpha is only transiently expressed in the cortex during neonatal development and is very low in uninjured adult cortex. Following MCAO, ERalpha mRNA expression is upregulated in the cortex of female rats, but the mechanism of this increase is still unknown. It is also unknown whether a similar increase in ERalpha expression in seen in males. In the following studies, male and vehicle or estradiol-treated ovariectomized (OVX) female rats underwent MCAO to investigate the regulation of ERalpha expression after ischemia. Twenty-four hours after surgery, mRNA or genomic DNA was collected from 1 mm micropunches taken from 300 mum brain sections for quantitative reverse transcription-polymerase chain reaction (RT-PCR) or methylation-specific (MSP) PCR, respectively. Additionally, adjacent 20 mum sections were processed for ERalpha immunohistochemistry. In OVX females, ERalpha mRNA and protein were increased in the ischemic cortex, but unchanged in males. We hypothesized that this increase in ERalpha in females is due to a reversal of gene silencing by DNA methylation. Using MSP targeting of CpG islands within the 5' untranslated region (UTR) of the rat ERalpha gene, we found that ischemia decreased methylation in the ischemic cortex of both groups of females, but there was no change in methylation in males. Using chromatin immunoprecipitation, we found that MeCP2 associates with ERalpha 5'UTR corresponding with the methylation status of the promoter. These data are the first to demonstrate a difference in the regulation of ERalpha expression in response to MCAO between males and females and that methylation of the ERalpha gene corresponds with mRNA levels in the brain.
Collapse
|
61
|
Alonso A, Moreno M, Ordóñez P, Fernández R, Pérez C, Díaz F, Navarro A, Tolivia J, González C. Chronic estradiol treatment improves brain homeostasis during aging in female rats. Endocrinology 2008; 149:57-72. [PMID: 17901235 DOI: 10.1210/en.2007-0627] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aging is associated with a reduction in metabolic function, insulin resistance, increased incidence of neurodegenerative diseases, and memory or cognitive dysfunction. In aging females, loss of gonadal function determines the beginning of the period of reduced metabolic function. Estrogens have neuroprotective effects, but the mechanisms by which they exert these effects remain unclear. The effects of estradiol treatment on the activation of the insulin receptor substrate (IRS)-1 signaling pathway, the interactions between estrogen receptor (ER)-alpha and IRS-1 and the p85alpha subunit of phosphatidylinositol-3 kinase, together with the possible effects of estradiol treatment on glucose transporter-3 and -4 levels, were investigated in female rats. The level of expression of each glucose transporter was greater in control and estradiol-treated groups than in the ovariectomized group. Interactions of ERalpha46-IRS-1, ERalpha46-p85alpha, and p85alpha-IRS-1, as well as IRS-1 phosphorylation, appeared to increase with estradiol treatment. The results indicate that estradiol treatment improves some aspects of neuronal homeostasis that are affected by aging; this may indicate that estradiol has neuroprotective effects in female rats. Additional animal studies are required to clarify the neuroprotective role of estradiol in relation to other important molecules involved in the IRS-1-phosphatidylinositol-3 kinase signaling pathway.
Collapse
Affiliation(s)
- Ana Alonso
- Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
González-Arenas A, Avendaño-Vázquez SE, Cabrera-Wrooman A, Tapia-Carrillo D, Larrea F, García-Becerra R, García-Sáinz JA. Regulation of LPA receptor function by estrogens. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:253-62. [PMID: 18166159 DOI: 10.1016/j.bbamcr.2007.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2007] [Revised: 11/18/2007] [Accepted: 11/20/2007] [Indexed: 01/01/2023]
Abstract
17beta-Estradiol induced LPA(1) receptor desensitization in C9 cells stably expressing LPA(1) receptors and transiently expressing estrogen receptor alpha. Such desensitization was evidenced by a reduction in lysophosphatidic acid-mediated Ca(2+)mobilization and it was associated to receptor phosphorylation and internalization. These effects of 17beta-estradiol were rapid (taking place over 5 min) and were blocked by the estrogen receptor antagonist ICI 182780. Similarly, inhibitors of phosphoinositide 3-kinase (wortmannin and LY294002) and of protein kinase C (staurosporine and Gö 6976) blocked 17beta-estradiol-induced LPA(1) receptor desensitization and phosphorylation. Confocal microscopy evidenced LPA(1) receptor internalization in response to 17beta-estradiol treatment. Association between LPA(1) receptors and protein kinase C alpha was suggested by co-immunoprecipitation assays. Protein kinase C alpha was associated with LPA(1) receptors in the absence of stimulus and such association further increased in a dynamic fashion in response to 17beta-estradiol. The results demonstrated that in C9 cells estrogens modulate LPA(1) action through estrogen receptor alpha with the participation of protein kinase C alpha and phosphoinositide 3-kinase.
Collapse
Affiliation(s)
- Aliesha González-Arenas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Apartado Postal 70-248, México, D. F. 04510. México
| | | | | | | | | | | | | |
Collapse
|
63
|
Kipp JL, Kilen SM, Woodruff TK, Mayo KE. Activin regulates estrogen receptor gene expression in the mouse ovary. J Biol Chem 2007; 282:36755-65. [PMID: 17951260 DOI: 10.1074/jbc.m705143200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activin, a member of the transforming growth factor-beta superfamily, is an important modulator of follicle-stimulating hormone synthesis and secretion in the pituitary and plays autocrine/paracrine roles in the regulation of ovarian follicle development. From a microarray study on mouse ovarian granulosa cells, we discovered that the estrogen receptor beta (ERbeta) is inducible by activin. We previously demonstrated that estrogen suppresses activin gene expression, suggesting a feedback relationship between these two follicle-regulating hormones. The purpose of this study was to investigate fully activin A regulation of ER expression. Real time reverse transcription-PCR assays on cultured granulosa cells showed that both ERalpha and ERbeta mRNAs were induced by activin A at 4, 12, and 24 h in a dose-responsive manner. Western blots confirmed an increase in their protein levels. Consistent with increased ERalpha and ERbeta expression, activin A stimulated estradiol-induced estrogen response element promoter activity. Activin A stimulation of ER expression was a direct effect at the level of gene transcription, as it was not abolished by cycloheximide but was abolished by actinomycin D, and in transfected granulosa cells activin A stimulated ERalpha promoter activity. To investigate the effect of activin in vivo and, thus, its biological significance, we examined ER expression in inhibin transgenic mice that have decreased activin expression and discovered that these mice had decreased ERalpha and ERbeta expression in the ovary. We also found that ER mRNA levels were decreased in Müllerian inhibiting substance promoter (MIS)-Smad2 dominant negative mice that have impaired activin signaling through Smad2, and small interfering RNAs targeting Smad2 or Smad3 suppressed ERalpha promoter activation, suggesting that Smad2 and Smad3 are involved in regulating ER levels. Therefore, this study reveals an important role for activin in inducing the expression of ERs in the mouse ovary and suggests important interplay between activin and estrogen signaling.
Collapse
Affiliation(s)
- Jingjing L Kipp
- Department of Biochemistry, Molecular Biology and Cell Biology, and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | |
Collapse
|
64
|
Katsu Y, Hinago M, Sone K, Urushitani H, Guillette LJ, Iguchi T. In vitro assessment of transcriptional activation of the estrogen and androgen receptors of mosquitofish, Gambusia affinis affinis. Mol Cell Endocrinol 2007; 276:10-7. [PMID: 17669586 DOI: 10.1016/j.mce.2007.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 06/11/2007] [Accepted: 06/18/2007] [Indexed: 11/22/2022]
Abstract
Sex-steroid hormones are essential for normal reproductive activity in both sexes. Estrogens are necessary for ovarian differentiation during a critical developmental stage in many vertebrates and promote the growth and differentiation of the female reproductive system. Androgens play essential roles in the development and functioning of the vertebrate male reproductive system as well as actively supporting spermatogenesis. Importantly, recent studies suggest that androgens and estrogens have important reproductive roles in both males and females. To understand the molecular mechanisms of estrogen and androgen actions and to evaluate estrogen and androgen receptor-ligand interactions in the mosquitofish, Gambusia affinis affinis, we used degenerate primer sets and PCR techniques to isolated DNA fragments encoding estrogen receptor alpha (ERalpha; ESR1), ERbeta1 (ERbeta1) and ERbeta2 from the ovary. Full-length mosquitofish ER (mfER) cDNAs were obtained using cDNA library screening and RACE techniques. Amino acid sequences of mfERs showed over-all homology of 46% (alpha versus beta1), 43% (alpha versus beta2), and 52% (beta1 versus beta2). We applied the ERE-luciferase reporter assay system to characterize these receptors. In this transient transfection assay system using mammalian cells, the mfER proteins displayed estrogen-dependent activation of transcription. In addition to ERs, the transactivation of mosquitofish ARs (mfARs) previously isolated by our group, were examined using an androgen-responsive MMTV-luciferase assay system. Mosquitofish ARs showed androgen-dependent activation of transcription from the MMTV promoter. These data provide a basic tool allowing future studies examining the receptor-ligand interactions and endocrine disrupting mechanisms in mosquitofish and also expands our knowledge of estrogen and androgen receptor evolution.
Collapse
Affiliation(s)
- Yoshinao Katsu
- Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | | | | | | | | | | |
Collapse
|
65
|
Baulieu EE, Robel P. Non-genomic mechanisms of action of steroid hormones. CIBA FOUNDATION SYMPOSIUM 2007; 191:24-37; discussion 37-42. [PMID: 8582201 DOI: 10.1002/9780470514757.ch3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sex steroid hormones are known to act through intracellular receptors and their cognate hormone response elements, located in the promoters of hormone-regulated genes. However, this classical mechanism of action cannot account for a variety of rapid effects of steroids (within seconds or minutes). In this review, non-genomic modes of target cell responses to sex steroids are described. The prototypical example is the resumption of meiosis in amphibian oocytes, triggered by progesterone at the plasma membrane level. Membrane effects of progesterone may also account for sperm maturation. Other membrane-mediated effects of steroids are reviewed. Whether a steroid hormone might elicit responses from a single cell through both genomic and membrane mechanisms remains an open question.
Collapse
Affiliation(s)
- E E Baulieu
- INSERM U33, 80 rue de Général Leclerc, Bicêtre, France
| | | |
Collapse
|
66
|
Derks NM, Roubos EW, Kozicz T. Presence of estrogen receptor beta in urocortin 1-neurons in the mouse non-preganglionic Edinger-Westphal nucleus. Gen Comp Endocrinol 2007; 153:228-34. [PMID: 17632108 DOI: 10.1016/j.ygcen.2007.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 03/21/2007] [Accepted: 04/01/2007] [Indexed: 11/17/2022]
Abstract
Adaptation to stress involves the activity of the hypothalamic-pituitary-adrenal (HPA-) axis. Urocortin 1 (Ucn1) coordinates responses to stressors. An increasing body of evidence suggests that such responses are sexually dimorphic and in females depend on the phase of the estrous cycle. Previously, in the non-preganglionic Edinger-Westphal nucleus (npEW), moderate immunostaining of the estrogen receptor alpha (ERalpha) was demonstrated, whereas estrogen receptor beta (ERbeta) was found to be more abundant. We have aimed at confirming the presence and identifying the type of ER in Ucn1-containing neurons in the npEW in the mouse, and at assessing whether the degree of Ucn1 mRNA expression is gender-related. Using immunocytochemistry, we could not demonstrate ERalpha-immunoreactivity in the npEW, but we did show a high density of ERbeta-immunopositive neurons in the npEW of both male and female mice. A majority of Ucn1-positive neurons showed ERbeta-immunoreactivity in their nuclei. In situ hybridization and RT-PCR did not reveal significant differences in both the number of neurons expressing Ucn1 mRNA and the strength of their Ucn1 mRNA expression. We will extend our gender comparison to other phases of the estrous cycle.
Collapse
Affiliation(s)
- Nicole M Derks
- Department of Cellular Animal Physiology, Integrative Physiology, EURON Graduate School for Neuroscience, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | | |
Collapse
|
67
|
Agras K, Willingham E, Shiroyanagi Y, Minasi P, Baskin LS. Estrogen receptor-alpha and beta are differentially distributed, expressed and activated in the fetal genital tubercle. J Urol 2007; 177:2386-92. [PMID: 17509364 DOI: 10.1016/j.juro.2007.01.111] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Indexed: 11/20/2022]
Abstract
PURPOSE We examined the ontogenic and sex specific expression of estrogen receptor-alpha and beta in mouse genital tubercles and assessed the effects of in utero estrogen exposure on these parameters. MATERIALS AND METHODS Expression of the 2 genes was detected in mouse genital tubercles from fetuses collected on gestational days 12, 14, 16 and 18, and from newborns using immunohistochemistry and quantitative polymerase chain reaction. Pregnant dams were exposed to ethinyl estradiol or corn oil as the control. RESULTS Estrogen receptor-alpha and beta proteins first appeared on gestational days 12 and 14, respectively. The 2 proteins were expressed in the urethral plate and mesenchyma. Staining intensity was more prominent in the mesenchyma for estrogen receptor-alpha and in the urethral plate for estrogen receptor-beta. Female genital tubercles expressed more estrogen receptor-alpha than male genital tubercles (p <0.01), while estrogen receptor-alpha expression increased gradually in the 2 sexes until birth. Estrogen receptor-beta expression did not differ between males and females, and it showed no notable variation during fetal life. Ethinyl estradiol led to a 2.1 and 3.8-fold increase in estrogen receptor-alpha expression in females and in males with hypospadias (p = 0.002 and 0.04, respectively). Estrogen receptor-beta expression did not change in response to ethinyl estradiol. CONCLUSIONS This study provides in vivo evidence that estrogen receptor-alpha expression in the genital tubercles of each sex increases until parturition but estrogen receptor-beta expression does not, implying genital tubercle sensitivity to estrogen increases during fetal life. Exogenous administration of estrogens results in a response of increased expression of estrogen receptor-alpha but not of estrogen receptor-beta. These differential findings for estrogen receptor-alpha and beta imply that the 2 receptors may have different roles in normal or anomalous genital tubercle development.
Collapse
Affiliation(s)
- Koray Agras
- Institute for the Study and Treatment of Hypospadias, Department of Urology, UCSF Children's Medical Center, University of California-San Francisco, San Francisco, CA, USA.
| | | | | | | | | |
Collapse
|
68
|
Matsumoto T, Nakamura AM, Mori K, Akiyama I, Hirose H, Takahashi Y. Oyster estrogen receptor: cDNA cloning and immunolocalization. Gen Comp Endocrinol 2007; 151:195-201. [PMID: 17324427 DOI: 10.1016/j.ygcen.2007.01.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 01/04/2007] [Accepted: 01/19/2007] [Indexed: 10/23/2022]
Abstract
A cDNA encoding the Pacific oyster, Crassostrea gigas, estrogen receptor (cgER) was cloned using degenerate PCR primers. The open reading frame predicted 485 amino acid residues. Comparisons of the amino acid sequence of cgER with other mollusk ERs show high similarities of the C domain (95-97%), and the E domain (56-66%). The amino acid sequence of the C domain of cgER shows 86 and 89% identity with the respective sequences of human ER-alpha and ER-beta. The amino acid sequence of the E domain of cgER shows 45% identity with those of human ER-alpha and ER-beta. The phylogenetic analysis indicated that the cgER is an ortholog of the other mollusk ERs. In the C domain, the positions of cysteine residues and other residues around them that constitute the two zinc finger motifs and the P-box are conserved. The cgER mRNA was expressed in various tissues including the ovary. Reporter gene assay revealed that cgER is unresponsive to estrogen. This result is similar to those of other mollusk ERs. ER immunoreactivity was localized mainly in the nuclei of follicle cells, the site of vitellogenin synthesis, and in oocytes. This result suggests that cgER could work as a nuclear receptor.
Collapse
Affiliation(s)
- Toshie Matsumoto
- National Research Institute of Aquaculture, Minami-ise, Mie 516-0193, Japan.
| | | | | | | | | | | |
Collapse
|
69
|
Dahlman-Wright K, Cavailles V, Fuqua SA, Jordan VC, Katzenellenbogen JA, Korach KS, Maggi A, Muramatsu M, Parker MG, Gustafsson JA. International Union of Pharmacology. LXIV. Estrogen receptors. Pharmacol Rev 2007; 58:773-81. [PMID: 17132854 DOI: 10.1124/pr.58.4.8] [Citation(s) in RCA: 373] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Hart SA, Snyder MA, Smejkalova T, Woolley CS. Estrogen mobilizes a subset of estrogen receptor-alpha-immunoreactive vesicles in inhibitory presynaptic boutons in hippocampal CA1. J Neurosci 2007; 27:2102-11. [PMID: 17314305 PMCID: PMC6673535 DOI: 10.1523/jneurosci.5436-06.2007] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2005] [Revised: 01/21/2007] [Accepted: 01/22/2007] [Indexed: 01/01/2023] Open
Abstract
Although the classical mechanism of estrogen action involves activation of nuclear transcription factor receptors, estrogen also has acute effects on neuronal signaling that occur too rapidly to involve gene expression. These rapid effects are likely to be mediated by extranuclear estrogen receptors associated with the plasma membrane and/or cytoplasmic organelles. Here we used a combination of serial-section electron microscopic immunocytochemistry, immunofluorescence, and Western blotting to show that estrogen receptor-alpha is associated with clusters of vesicles in perisomatic inhibitory boutons in hippocampal CA1 and that estrogen treatment mobilizes these vesicle clusters toward synapses. Estrogen receptor-alpha is present in approximately one-third of perisomatic inhibitory boutons, and specifically in those that express cholecystokinin, not parvalbumin. We also found a high degree of extranuclear estrogen receptor-alpha colocalization with neuropeptide Y. Our results suggest a novel mode of estrogen action in which a subset of vesicles within a specific population of inhibitory boutons responds directly to estrogen by moving toward synapses. The mobilization of these vesicles may influence acute effects of estrogen mediated by estrogen receptor-alpha signaling at inhibitory synapses.
Collapse
Affiliation(s)
- Sharron A. Hart
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208
| | - Melissa A. Snyder
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208
| | - Tereza Smejkalova
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208
| | - Catherine S. Woolley
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
71
|
Powell SA, Smith BB, Timm KI, Menino AR. Expression of estrogen receptors α and β in the corpus luteum and uterus from non-pregnant and pregnant llamas. Mol Reprod Dev 2007; 74:1043-52. [PMID: 17219432 DOI: 10.1002/mrd.20684] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Because estrogen may be involved in maternal recognition of pregnancy and embryonic migration in llamas, expression of estrogen receptor subtypes alpha (ERalpha) and beta (ERbeta) was evaluated in corpus luteum (CL), endometrium, and uterus using relative RT-PCR. Tissues were recovered from sterile-mated (SM) and pregnant (PG) females during Days 7-11 and 7-13 (Day 0 = day of mating), respectively, and follicular phase and juvenile females. Luteal expression of ERalpha and beta was similar (P > 0.10) in SM and PG females and within Days 7-11, however, expression of ERalpha in ovarian tissue from follicular phase females was greater (P < 0.05) than Days 7 and 9 CL. Uterus expressed less ERalpha and beta compared to endometrium (P = 0.07 and P < 0.01, respectively). Expression of ERalpha was greater (P < 0.05) in Day 7 and follicular phase uteri than Days 9 and 11, Day 13 PG and juvenile uteri. Uterine ERbeta expression was greater (P = 0.09) in PG versus SM females and in mated compared to follicular phase females (P < 0.05). Endometrial expression of ERalpha and beta did not differ (P > 0.10) between SM and PG females or by day. The presence of luteal ER during this period may mean a role for estradiol in maternal recognition of pregnancy. Observed increases in uterine ER expression with no changes in endometrium suggest expression increased in myometrium and/or perimetrium. Upregulation of myometrial ERbeta in PG females may be involved in supporting uterine migration of the embryo.
Collapse
Affiliation(s)
- Susan A Powell
- Department of Animal Sciences, Oregon State University, Corvallis, OR 97331-6702, USA
| | | | | | | |
Collapse
|
72
|
Alonso A, Fernández R, Ordóñez P, Moreno M, Patterson AM, González C. Regulation of estrogen receptor alpha by estradiol in pregnant and estradiol treated rats. Steroids 2006; 71:1052-61. [PMID: 17030051 DOI: 10.1016/j.steroids.2006.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 07/07/2006] [Accepted: 09/05/2006] [Indexed: 10/24/2022]
Abstract
Estrogens play an important role in tissue metabolism through specific regulation of several intracellular pathways. We studied ERalpha regulation in muscle and adipose tissue from pregnant and estradiol treated rats. In both groups, we identified three different ERalpha inmunoreactive proteins (80, 67 and 46 kDa) using total protein extracts. Because it has been showed that estrogens are able to promote rapid effects in several cellular models, we looked for three ERalpha-related proteins at plasma membrane. In skeletal muscle of both groups, we positively identified the three ERalpha-related isoforms in plasma membrane, but in adipose tissue from pregnant we were not able to identify ERalpha67, and in estradiol treated animals ERalpha80 was absent. Taking together, our results showed a tissue-specific regulation of whole-cell ERalpha-related proteins and ERalpha located at plasma membrane, which should be involved in non-genomic actions of 17beta-estradiol. The role of the three ERalpha inmunoreactive proteins is unknown, however, seems probably related to rapid activation of signalling pathways.
Collapse
Affiliation(s)
- Ana Alonso
- Department of Functional Biology. Physiology Area, University of Oviedo, C/ Julián Clavería s/n, 33006 Oviedo, Spain
| | | | | | | | | | | |
Collapse
|
73
|
Alvaro D, Barbaro B, Franchitto A, Onori P, Glaser SS, Alpini G, Francis H, Marucci L, Sterpetti P, Ginanni-Corradini S, Onetti Muda A, Dostal DE, De Santis A, Attili AF, Benedetti A, Gaudio E. Estrogens and insulin-like growth factor 1 modulate neoplastic cell growth in human cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:877-88. [PMID: 16936263 PMCID: PMC1698823 DOI: 10.2353/ajpath.2006.050464] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We investigated the expression of estrogen receptors (ERs), insulin-like growth factor 1 (IGF-1), and IGF-1R (receptor) in human cholangiocarcinoma and cholangiocarcinoma cell lines (HuH-28, TFK-1, Mz-ChA-1), evaluating the role of estrogens and IGF-1 in the modulation of neoplastic cell growth. ER-alpha, ER-beta, IGF-1, and IGF-1R were expressed (immunohistochemistry) in all biopsies (18 of 18) of intrahepatic cholangiocarcinoma. ER-alpha was expressed (Western blot) only by the HuH-28 cell line (intrahepatic cholangiocarcinoma), whereas ER-beta, IGF-1, and IGF-1R were expressed in the three cell lines examined. In serum-deprived HuH-28 cells, serum readmission induced stimulation of cell proliferation that was inhibited by ER and IGF-1R antagonists. 17beta-Estradiol and IGF-1 stimulated proliferation of HuH-28 cells to a similar extent to that of MCF7 (breast cancer) but greater than that of TFK-1 and Mz-ChA-1, inhibiting apoptosis and exerting additive effects. These effects of 17beta-estradiol and IGF-1 were associated with enhanced protein expression of ER-alpha, phosphorylated (p)-ERK1/2 and pAKT but with decreased expression of ER-beta. Finally, transfection of IGF-1R anti-sense oligonucleotides in HuH-28 cells markedly decreased cell proliferation. In conclusion, human intrahepatic cholangiocarcinomas express receptors for estrogens and IGF-1, which cooperate in the modulation of cell growth and apoptosis. Modulation of ER and IGF-1R could represent a strategy for the management of cholangiocarcinoma.
Collapse
Affiliation(s)
- Domenico Alvaro
- Department of Clinical Medicine, Division of Gastroenterology, University of Rome, via R. Rossellini 51, 00137 Rome, Italy. domenico.alvaro@uniroma1
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Yoshino M, Mizutani T, Yamada K, Yazawa T, Ogata-Kawata H, Sekiguchi T, Kajitani T, Miyamoto K. Co-activator p120 is increased by gonadotropins in the rat ovary and enhances progesterone receptor activity. Reprod Biol Endocrinol 2006; 4:50. [PMID: 17014737 PMCID: PMC1617106 DOI: 10.1186/1477-7827-4-50] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Accepted: 10/03/2006] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Ovarian follicular development is primarily dependent on pituitary gonadotropins. Identification of gonadotropin-inducible genes in the ovary is one of the effective approaches for the study of follicular development. In this study we identify rat homologue of p120, a nuclear transcription co-activator, as one of the FSH inducible genes in the rat granulosa cells. METHODS A full-length cDNA encoding rat p120 was cloned, and expression of the gene in the ovary was examined by Northern blotting. Tissue localization of p120 was examined by in situ hybridization. Cellular functions of p120 were studied by co-transfection of rat p120 gene together with estrogen receptor (ER)-alpha, ER-beta, androgen receptor (AR), or progesterone receptor (PR) genes. RESULTS A full-length cDNA encoding rat p120 was characterized as a protein with 957 amino acid residues. Rat p120 was expressed ubiquitously, but strongly in the ovary and the testis. Expression of p120 mRNA was also induced in vivo by PMSG or PMSG/hCG treatment. Strong expression of p120 mRNA was observed in the granulosa cells of pre-ovulatory large antral follicles. Progesterone receptor was co-localized with p120 in the large antral follicles. Co-transfection experiments revealed that rat p120 activated AR, ER-alpha, ER-beta, and PR in the presence of their respective ligands. CONCLUSION These observations suggest that rat p120 is strongly induced in the ovarian granulosa cells, and may work together with PR in the granulosa cells of ovulatory follicles to promote the ovulation process.
Collapse
Affiliation(s)
- Miki Yoshino
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Tetsuya Mizutani
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Kazuya Yamada
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Takashi Yazawa
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Hiroko Ogata-Kawata
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Toshio Sekiguchi
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Takashi Kajitani
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| | - Kaoru Miyamoto
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Shimoaizuki, Matsuoka, Fukui 910-1193, Japan
- CREST, JST (Japan Science and Technology), Japan
| |
Collapse
|
75
|
Katsu Y, Kohno S, Oka T, Mitsui N, Tooi O, Santo N, Urushitani H, Fukumoto Y, Kuwabara K, Ashikaga K, Minami S, Kato S, Ohta Y, Guillette LJ, Iguchi T. Molecular cloning of estrogen receptor alpha (ERalpha; ESR1) of the Japanese giant salamander, Andrias japonicus. Mol Cell Endocrinol 2006; 257-258:84-94. [PMID: 16919388 DOI: 10.1016/j.mce.2006.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Revised: 07/11/2006] [Accepted: 07/13/2006] [Indexed: 11/16/2022]
Abstract
Estrogens are essential for normal reproductive activity in females and males and for ovarian differentiation during a critical developmental stage in many vertebrates. To understand the molecular mechanisms of estrogen action and to evaluate estrogen receptor ligand interactions in the Japanese giant salamander (Andrias japonicus), we isolated cDNA encoding the estrogen receptor (ER) from the liver. A full-length Japanese giant salamander ER cDNA (jgsER) was obtained using 5' and 3' rapid amplification cDNA ends (RACE). The deduced amino acid sequence of the jgsER showed high identity to the Xenopus ERalpha (ESR1) (77.7%). We have applied both the conventional ERE-luciferase reporter assay system and the GAL4-transactivation system to characterize this receptor. In two different transient transfection assay systems using mammalian cells, the jgsER protein displayed estrogen-dependent activation of transcription. The GAL4-transactivation system showed about 10-fold greater activity of the estrogen receptor by hormone when compared to the conventional ERE-luciferase reporter assay system. Tissue distribution of ERalpha mRNA was examined and kidney, ovary and liver exhibited expression. This is the first isolation of an estrogen receptor from a salamander and also is the first functional cDNA obtained from the Japanese giant salamander, an endangered species considered a special natural monument of Japan.
Collapse
Affiliation(s)
- Yoshinao Katsu
- Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Ascenzi P, Bocedi A, Marino M. Structure-function relationship of estrogen receptor alpha and beta: impact on human health. Mol Aspects Med 2006; 27:299-402. [PMID: 16914190 DOI: 10.1016/j.mam.2006.07.001] [Citation(s) in RCA: 361] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
17Beta-estradiol (E2) controls many aspects of human physiology, including development, reproduction and homeostasis, through regulation of the transcriptional activity of its cognate receptors (ERs). The crystal structures of ERs with agonists and antagonists and the use of transgenic animals have revealed much about how hormone binding influences ER conformation(s) and how this conformation(s), in turn, influences the interaction of ERs with co-activators or co-repressors and hence determines ER binding to DNA and cellular outcomes. This information has helped to shed light on the connection between E2 and the development or progression of numerous diseases. Current therapeutic strategy in the treatment of E2-related pathologies relies on the modulation of ER trancriptional activity by anti-estrogens; however, data accumulated during the last five years reveal that ER activities are not only restricted to the nucleus. ERs are very mobile proteins continuously shuttling between protein targets located within various cellular compartments (e.g., membrane, nucleus). This allows E2 to generate different and synergic signal transduction pathways (i.e., non-genomic and genomic) which provide plasticity for cell response to E2. Understanding the structural basis and the molecular mechanisms by which ER transduce E2 signals in target cells will allow to create new pharmacologic therapies aimed at the treatment of a variety of human diseases affecting the cardiovascular system, the reproductive system, the skeletal system, the nervous system, the mammary gland, and many others.
Collapse
Affiliation(s)
- Paolo Ascenzi
- Department of Biology, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | | | | |
Collapse
|
77
|
González-Arenas A, Aguilar-Maldonado B, Avendaño-Vázquez SE, García-Sáinz JA. Estrogens Cross-Talk to α1b-Adrenergic Receptors. Mol Pharmacol 2006; 70:154-62. [PMID: 16638969 DOI: 10.1124/mol.106.025064] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
beta-Estradiol induced alpha1b-adrenergic receptor desensitization in U373 MG cells stably expressing alpha1b-adrenoceptors, as evidenced by a reduction in the adrenergic-mediated Ca2+ mobilization; desensitization was associated with receptor phosphorylation and internalization. These effects of beta-estradiol were rapid (taking place during 15 min) and were blocked by the estrogen receptor antagonist ICI 182,780 (faslodex). Likewise, inhibitors of phosphoinositide 3-kinase [wortmannin and 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002)] and of protein kinase C [staurosporine, 3-[1-[3-(amidinothio)propyl-1H-indol-3-yl]-3-(1-methyl-1H-indol-3-yl) maleimide (Ro31-8220), and rottlerin] blocked the desensitization and phosphorylation of alpha1b-adrenoceptors induced by estradiol. The formation of a complex was suggested by coimmunoprecipitation assays. The regulatory and catalytic subunits of phosphoinositide 3-kinase (p85 and p110) and protein kinase C delta were associated with alpha1b-adrenoceptors in the absence of stimulus, and such association further increased in a dynamic fashion in response to beta-estradiol. In cells cotransfected with the estrogen receptor alpha and alpha1b-adrenoceptors, beta-estradiol induced phosphorylation, desensitization and internalization of the adrenergic receptors; pretreatment with ICI 182,780 inhibited these effects. Our data support the idea that estrogens modulate alpha1b-adrenergic action through estrogen receptor alpha.
Collapse
|
78
|
Shi SF, Yu CQ. [Progress in research on phytoestrogens and their effect targets]. ACTA ACUST UNITED AC 2006; 3:408-10. [PMID: 16159582 DOI: 10.3736/jcim20050521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shu-Fang Shi
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | | |
Collapse
|
79
|
Wilson ME, Dimayuga FO, Reed JL, Curry TE, Anderson CF, Nath A, Bruce-Keller AJ. Immune modulation by estrogens: role in CNS HIV-1 infection. Endocrine 2006; 29:289-97. [PMID: 16785604 DOI: 10.1385/endo:29:2:289] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 10/24/2005] [Accepted: 10/24/2005] [Indexed: 11/11/2022]
Abstract
Experimental and epidemiological data suggest that estrogen can be protective in both brain injury and infection. While estrogens can act directly on neurons to promote neuronal survival, estrogen also has antiinflammatory properties that may contribute to overall neuroprotection. Accordingly, estrogens may have particular relevance in chronic neuroimmune disorders such as HIV dementia. As AIDS is now a leading cause of death among women in their reproductive years, understanding the role that female sex hormones might play in the physiology of HIV-1 infection is especially critical. Indeed, there is accumulating evidence that many manifestations of HIV differ in women. For instance, it is now well established that women present with a lower viral titer at the time of seroconversion, have lower HIV viral loads compared to men at similar stages of disease, and may have altered disease progression during pregnancy. Conversely, while epidemiological studies suggest that women may be more vulnerable to certain late-stage AIDS-related illnesses including HIV dementia, there is accumulating data that strongly suggest an estrogen-deficient state is associated with long-term HIV infection in some women. Evaluated as a whole, existing evidence indicates that estrogen can directly protect neurons from damage, can modulate brain inflammation, and could act to maintain low titers of the HIV-1 virus. Accordingly, it can be hypothesized that maintenance of serum estradiol levels could decrease the incidence of HIV dementia and other AIDS-related neurological syndromes in HIV-1 positive women. In this article, we both summarize current understanding and present new data related to the potential mechanisms whereby estrogen could modulate the mechanics and the consequences of HIV-1 infection in the brain and thereby thwart the development of HIV dementia.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
| | | | | | | | | | | | | |
Collapse
|
80
|
Isgor C, Watson SJ. Estrogen receptor alpha and beta mRNA expressions by proliferating and differentiating cells in the adult rat dentate gyrus and subventricular zone. Neuroscience 2005; 134:847-56. [PMID: 15994024 DOI: 10.1016/j.neuroscience.2005.05.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Revised: 04/19/2005] [Accepted: 05/11/2005] [Indexed: 11/18/2022]
Abstract
Numerous factors modulate neurogenesis in the adult dentate gyrus and subventricular zone, but it is often not clear if the modulation is mediated by direct effects on the proliferating and differentiating cells or secondary to effects on other cells. Also, while some factors selectively affect neurogenesis in one of the neurogenetic zones, it is not clear how selectivity is achieved. Estrogen is a hormonal modulator of neurogenesis. To address the issues of direct versus indirect control and regional specificity we investigated the colocalization of immunoreactivity for a proliferating cell marker, Ki-67, and a marker for migrating and differentiating cells with a neuronal phenotype, doublecortin, with the expressions of mRNA for estrogen receptors alpha and beta. We found an extensive colocalization of estrogen receptor alpha with both markers in the dentate gyrus and only with Ki-67 in the subventricular zone. An extensive colocalization of estrogen receptor beta with both markers was found in the dentate gyrus, but only a few Ki-67-immunoreactive and no doublecortin-immunoreactive cells of the subventricular zone expressed estrogen receptor beta mRNA. Estrogen receptor alpha and beta mRNAs were not expressed in other telencephalic Ki-67-immunoreactive cells or in constitutively doublecortin-immunoreactive cells of the piriform cortex. The extensive colocalization of immunoreactive markers for cell proliferation and differentiation with mRNAs for estrogen receptor alpha and estrogen receptor beta points to the direct modulation of dentate cell proliferation, differentiation and survival by estrogen, while direct effects of estrogen in the subventricular zone appear restricted to estrogen receptor alpha-mediated effects operating at the time of cell proliferation.
Collapse
Affiliation(s)
- C Isgor
- Department of Biomedical Science, Charles E. Schmidt Biomedical Center, Florida Atlantic University, Boca Raton, FL 33431-0991, USA.
| | | |
Collapse
|
81
|
Oropeza MV, Orozco S, Ponce H, Campos MG. Tofupill lacks peripheral estrogen-like actions in the rat reproductive tract. Reprod Toxicol 2005; 20:261-6. [PMID: 15878261 DOI: 10.1016/j.reprotox.2005.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Revised: 01/16/2005] [Accepted: 02/14/2005] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The objective of this study was to evaluate the estrogenic effect of phytoestrogens contained in a commercial food supplement (Tofupill) on the reproductive tract of ovariectomized rats. METHODS Food supplement (3.4 or 10.2 mg/kg) and conjugated equine estrogens (CEE, 31 or 100 microg/kg) were orally administered, daily during 14 days to ovariectomized rats. At the end of treatment, the following determinations were done: dry and wet uterine weight, vaginal epithelium condition, and uterine serotonin-induced contractile response. A group treated with 17beta-estradiol was included as control for serotonin-induced contractile response. RESULTS Food supplement did not display clear estrogenic effects on vaginal epithelium, uterine weight or myometrial sensitivity to serotonin, whereas high doses of conjugated equine estrogens showed estrogenic action. CONCLUSIONS The present data showed that Tofupill displayed a lower estrogenic effect than conjugated equine estrogens, which are one of the most commonly used hormone replacement therapy for postmenopausal women. However, further studies are needed to evaluate the risk associated to the use of Tofupill as an alternative to hormone replacement therapy for postmenopausal women.
Collapse
Affiliation(s)
- Martha V Oropeza
- Medical Research Unit in Pharmacology, National Medical Center SXXI, IMSS, Mexico City, Mexico
| | | | | | | |
Collapse
|
82
|
Ottem EN, Godwin JG, Krishnan S, Petersen SL. Dual-phenotype GABA/glutamate neurons in adult preoptic area: sexual dimorphism and function. J Neurosci 2005; 24:8097-105. [PMID: 15371511 PMCID: PMC6729791 DOI: 10.1523/jneurosci.2267-04.2004] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
It is generally assumed that the inhibitory neurotransmitter GABA and the stimulatory neurotransmitter glutamate are released from different neurons in adults. However, this tenet has made it difficult to explain how the same afferent signals can cause opposite changes in GABA and glutamate release. Such reciprocal release is a central mechanism in the neural control of many physiological processes including activation of gonadotropin-releasing hormone (GnRH) neurons, the neural signal for ovulation. Activation of GnRH neurons requires simultaneous suppression of GABA and stimulation of glutamate release, each of which occurs in response to a daily photoperiodic signal, but only in the presence of estradiol (E2). In rodents, E2 and photoperiodic signals converge in the anteroventral periventricular nucleus (AVPV), but it is unclear how these signals differentially regulate GABA and glutamate secretion. We now report that nearly all neurons in the AVPV of female rats express both vesicular glutamate transporter 2 (VGLUT2), a marker of hypothalamic glutamatergic neurons, as well as glutamic acid decarboxylase and vesicular GABA transporter (VGAT), markers of GABAergic neurons. These dual-phenotype neurons are the main targets of E2 in the region and are more than twice as numerous in females as in males. Moreover, dual-phenotype synaptic terminals contact GnRH neurons, and at the time of the surge, VGAT-containing vesicles decrease and VGLUT2-containing vesicles increase in these terminals. Thus, we propose a new model for ovulation that includes dual-phenotype GABA/glutamate neurons as central transducers of hormonal and neural signals to GnRH neurons.
Collapse
Affiliation(s)
- Erich N Ottem
- Department of Biology, Center for Neuroendocrine Studies, University of Massachusetts-Amherst, Amherst, Massachusetts 01002, USA
| | | | | | | |
Collapse
|
83
|
Shimizu T, Kamegai J, Tamura H, Ishii S, Sugihara H, Oikawa S. The estrogen receptor (ER) α, but not ER β, gene is expressed in hypothalamic growth hormone-releasing hormone neurons of the adult female rat. Neurosci Res 2005; 52:121-5. [PMID: 15811559 DOI: 10.1016/j.neures.2005.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Revised: 02/02/2005] [Accepted: 02/04/2005] [Indexed: 10/25/2022]
Abstract
Growth hormone (GH) synthesis and release from pituitary somatotropes is controlled by the opposing actions of the hypothalamic neuropeptides, GH-releasing hormone (GHRH) in the arcuate nucleus (ARC), and somatostatin in the periventricular nucleus (PeV) and ARC. There is a striking sex difference in the pattern of GH secretion in rats. We have previously demonstrated in male rats that 70% of GHRH neurons in the ARC contain the estrogen receptor alpha (ER alpha) gene, whereas less than 5% of somatostatin neurons in the ARC and PeV expressed the ER alpha or ER beta gene. In addition, it has been reported that the PeV somatostatin neurons of neither sex possess ER immunoreactivity. However, there is no available data about colocalization of ERs and GHRH and/or somatostatin in the ARC of female rats. In this study, we used in situ hybridization in the adult female rat brain to determine whether GHRH neurons and/or somatostatin neurons in the ARC coexpress the ER alpha or ER beta gene. In the ARC, ER alpha mRNA was seen in the ventrolateral region where GHRH mRNA signals were also observed, and in the dorsomedial region where somatostatin mRNA signals were also observed. From studies using adjacent sections through these areas, the distribution of these cells appeared to overlap in part with that of cells containing ER alpha mRNA. On the other hand, few positive cells for ER beta mRNA were observed in the ARC. The double-label in situ hybridization studies showed that in the ARC, 73.4% of GHRH neurons contain ER alpha mRNA, whereas less than 5% of somatostatin neurons express the ER alpha gene. These results indicated that the majority of the GHRH neurons in ARC have ER alpha, but not ER beta, and few somatostatin neurons in ARC have ER alpha or ER beta in either adult female or male rats, suggesting that colocalization with ERs in GHRH and/or somatostatin neurons is not an important determinant of the gender specific pattern of GH secretion.
Collapse
Affiliation(s)
- Takako Shimizu
- Department of Medicine, Nippon Medical School, 1-1-5 Sendagi, Tokyo 113-8603, Japan.
| | | | | | | | | | | |
Collapse
|
84
|
Adachi T, Yasuda K, Mori C, Yoshinaga M, Aoki N, Tsujimoto G, Tsuda K. Promoting insulin secretion in pancreatic islets by means of bisphenol A and nonylphenol via intracellular estrogen receptors. Food Chem Toxicol 2005; 43:713-9. [PMID: 15778011 DOI: 10.1016/j.fct.2005.01.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Revised: 01/07/2005] [Accepted: 01/19/2005] [Indexed: 11/29/2022]
Abstract
In this study, we investigated the effects of endocrine disrupters bisphenol A (BPA) and nonylphenol (NP) on insulin secretion from rat pancreatic islets. Following acute exposure to BPA and NP, neither BPA nor NP (0.1, 1, 10, 100 and 1000 microg/l) affected insulin secretion in concentrations of 16.7 mM glucose. However, insulin secretion following long-term exposure to BPA or NP for 24 h in 16.7 mM glucose was significantly higher than without exposure. To determine whether increased insulin secretion resulting from long-term exposure to BPA and NP is induced via intracellular estrogen receptors, we blocked the cytosolic/nuclear estrogen receptors, using actinomycin-D (Act-D), an inhibitor of RNA synthesis, and ICI 182,780 (ICI), an estrogen receptor inhibitor. Following long-term exposure to BPA (10 microg/l) or NP (10 microg/l), Act-D or ICI treatment eliminated the facilitation of insulin secretion. In conclusion, we have demonstrated for the first time that long-term exposure to endocrine disrupters, such as BPA and NP, promotes in vitro insulin secretion from the pancreatic islets, via cytosolic/nuclear estrogen receptors.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
85
|
Byers MJ, Zangl A, Phernetton TM, Lopez G, Chen DB, Magness RR. Endothelial vasodilator production by ovine uterine and systemic arteries: ovarian steroid and pregnancy control of ERalpha and ERbeta levels. J Physiol 2005; 565:85-99. [PMID: 15774511 PMCID: PMC1464491 DOI: 10.1113/jphysiol.2005.085753] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Pregnancy and the follicular phase are physiological states of elevated oestrogen levels and rises in uterine blood flow (UBF). The dramatic increase in utero-placental blood flow during gestation is required for normal fetal growth and development. Oestrogen exerts its vasodilatory effect by binding to its specific oestrogen receptors (ER) in target cells, resulting in increased expression and activity of endothelial nitric oxide synthase (eNOS) to relax vascular smooth muscle (VSM). However, the regulation of endothelial versus VSM ERalpha and ERbeta expression in uterine arteries (UAs) during the ovarian cycle, pregnancy and with exogenous hormone replacement therapy (HRT) are currently unknown. ER mRNA and protein localization was determined by in situ hybridization (ISH) using 35S-labelled riboprobes and immunohistochemistry (IHC), respectively. UA endothelial (UAendo), UA VSM, omental artery endothelium (OA endo), and OA VSM proteins were isolated and ERalpha and ERbeta protein expression was determined by Western analysis. We observed by ISH and IHC that ERalpha and ERbeta mRNA and protein were localized in both UAendo and UA VSM. Immunoblot data demonstrated ovarian hormone specific regulation of ERalpha and ERbeta protein in UAendo and UA VSM. Compared to luteal phase sheep, both ERalpha and ERbeta levels in UAendo were elevated in follicular phase sheep. Whereas ERbeta was elevated by pregnancy in UAendo and UA VSM, ERalpha was not appreciably altered. eNOS was increased in UAendo from follicular and pregnant sheep. Ovariectomized ewes (OVEX) had substantially reduced UAendo ERbeta, but not UAendo ERalpha or OAendo ERalpha and ERbeta. In contrast, OVEX increased UA VSM ERalpha and ERbeta and decreased OA VSM ERalpha and ERbeta. Treatment with oestradiol-17beta (E2beta), but not progesterone or their combination, increased UAendo ERalpha levels. The reduced ERbeta in UAendo from OVEX ewes was reversed by E(2)beta and progesterone treatment. While ERalpha and eNOS were not elevated in any other reproductive or non-reproductive endothelia tested, ERbeta was augmented by pregnancy in uterine, mammary, placenta, and coronary artery endothelia. ERalpha and ERbeta mRNA and protein are expressed in UA endothelium with expression levels depending on the endocrine status of the animal, indicating UA endothelium is a target for oestrogen action in vivo, and that the two receptors appear to be differentially regulated in a spatial and temporal fashion with regard to the reproductive status or HRT.
Collapse
Affiliation(s)
- Michael J Byers
- Perinatal Research Laboratories, University of Wisconsin-Madison Medical School, Department of Obstetrics and Gynecology, Atrium-B Meriter Hospital/Park , Madison, WI 53715, USA
| | | | | | | | | | | |
Collapse
|
86
|
Okada A, Sato T, Ohta Y, Iguchi T. SEX STEROID HORMONE RECEPTORS IN THE DEVELOPING FEMALE REPRODUCTIVE TRACT OF LABORATORY RODENTS. J Toxicol Sci 2005; 30:75-89. [PMID: 15928456 DOI: 10.2131/jts.30.75] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many chemicals released into the environment potentially disrupt the endocrine system in wildlife and humans. Some of these chemicals exhibit estrogenic activity by binding to the estrogen receptors. The developing organism is particularly sensitive to estrogenic chemicals during the critical period in which the induction of long-term changes and persistent molecular alterations in female reproductive tracts occur. Perinatal mouse and rat models can be utilized as indicators for determining the consequences of exposure to exogenous estrogenic agents, including possible xenoestrogens or environmental endocrine disruptors. Estrogen receptors (ER) and estrogen responsive genes, therefore, need to be identified in order to understand the molecular basis of estrogenic actions. Recent identifications of ER subtypes and isoforms make understanding target organ responses to these estrogenic chemicals even more difficult. Indeed, many reports suggest that these chemicals do affect the reproductive and developmental processes of female laboratory rodents that had been perinatally exposed, and that interactions between sex steroid hormone receptors occur. Much information concerning the expression of sex steroid receptors in rodents has been reported concerning the normal development of the Müllerian duct. Thus, accumulated information on the expression of ER subtypes and isoforms as well as that of progesterone and androgen receptors in laboratory rodents is herein reviewed, in addition to the presentation of our own data.
Collapse
Affiliation(s)
- Akinobu Okada
- Safety Research Laboratories, Institute for Drug Discovery Research, Yamanouchi Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | | | | |
Collapse
|
87
|
Kennedy AM, Shogren KL, Zhang M, Turner RT, Spelsberg TC, Maran A. 17beta-estradiol-dependent activation of signal transducer and activator of transcription-1 in human fetal osteoblasts is dependent on Src kinase activity. Endocrinology 2005; 146:201-7. [PMID: 15471961 DOI: 10.1210/en.2004-0486] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Estrogen is essential for normal growth and remodeling of bone. Although the mechanism of estrogen action on bone cells has been widely investigated, the full spectrum of signal transduction pathways activated by estrogen is unknown. In this report, we investigate the effects of the gonadal hormone 17beta-estradiol on the regulation of signal transducer and activator of transcription-1 (Stat1) protein in cultured human fetal osteoblast cells, devoid of the classical estrogen receptors (ERs). 17beta-estradiol (10 nM) led to rapid (within 15 min) activation of Stat1 protein as indicated by increases in tyrosine phosphorylation and DNA binding activity. Also, 17beta-estradiol increased gamma-activated sequence-dependent transcription in transient transfection assays, suggesting an increase in Stat protein-dependent transcription. Estrogen-dependent Stat1 activation was blocked in cells that transiently express dominant-negative Stat1 mutant protein. Activation of Stat1 by 17beta-estradiol was not inhibited by ER antagonist ICI 182,780, providing further evidence that it is not dependent on classical ERs. 17beta-Estradiol induced rapid (within 15 min) Stat1 phosphorylation and stimulated gamma-activated sequence-dependent transcription in ER-negative breast cancer cells, indicating that these results are not unique to bone cells. The rapid estrogenic effect involving the phosphorylation and activation of Stat1 was blocked in the presence of Src family kinase inhibitor PP2; activated Stat1 was associated with Src protein in estrogen-treated cells. These findings indicate the requirement for Src kinase pathways in estrogen-mediated Stat1 activation. Thus, the ER-independent activation of Stat1 in 17beta-estradiol-treated osteoblast and breast cancer cells may partially mediate the actions of estrogen on target cells.
Collapse
Affiliation(s)
- Angela M Kennedy
- Department of Orthopedics, Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | | | |
Collapse
|
88
|
Kanno S, Hirano S, Kayama F. Effects of the phytoestrogen coumestrol on RANK-ligand-induced differentiation of osteoclasts. Toxicology 2004; 203:211-20. [PMID: 15363596 DOI: 10.1016/j.tox.2004.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Revised: 05/28/2004] [Accepted: 06/07/2004] [Indexed: 11/23/2022]
Abstract
Phytoestrogens, which have structural similarity to 17beta-estradiol, have been reported to act as agonists/antagonists of estrogen in animals and humans. Estrogen is known to have an important role in maintaining bone mass, because the concentration of serum estrogen decreases after menopause and the estrogen deficiency causes bone loss. In this study, we investigated the effects of coumestrol and other phytoestrogens on osteoclast differentiation using estrogen receptor alpha-transfected RAW264.7 (RAW264.7-ERalpha) cells. When the cells were cultured with the receptor activator of nuclear factor kappa B-ligand (RANKL), both formation of tartrate-resistant acid phosphatase (TRAP) positive multinucleated cells and TRAP activity were increased compared with control cells that were cultured in the absence of RANKL. Coumestrol decreased RANKL-induced formation of TRAP-positive multinucleated cells and TRAP activity dose-dependently. RANKL-stimulated RAW264.7-ERalpha cells formed resorption pits on calcium phosphate films and the pit formation was inhibited by coumestrol in a dose-dependent manner. RT-PCR analyses revealed that coumestrol (10 microM) decreased mRNA levels of calcitonin receptor (CTR) and matrix metalloproteinase-9 (MMP9) in RANKL-treated cells. In addition, pretreatment of coumestrol decreased RANKL-induced phosphorylation of extracellular signal-regulated kinases/p44/42 (ERK1/2). These results suggest that coumestrol has an inhibitory effect on the differentiation of osteoclasts, at least partially via ERK1/2 pathway.
Collapse
Affiliation(s)
- Sanae Kanno
- Environmental Health Sciences Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan.
| | | | | |
Collapse
|
89
|
Perez S, Sendera TJ, Kordower JH, Mufson EJ. Estrogen receptor alpha containing neurons in the monkey forebrain: lack of association with calcium binding proteins and choline acetyltransferase. Brain Res 2004; 1019:55-63. [PMID: 15306238 DOI: 10.1016/j.brainres.2004.05.101] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2004] [Indexed: 11/29/2022]
Abstract
The present study used single and dual immunohistochemistry to determine the topography and chemical phenotype of ERalpha containing neurons within the monkey forebrain utilizing antibodies directed against the full-length human ERalpha (NCL-ER-6F11), calcium-binding proteins calbindin-D(28k), and parvalbumin as well as choline acetyltransferase (ChAT). Our findings demonstrate for the first time ERalpha immunoreactive (-ir) cells in the monkey cerebral cortex (layers I-II) and in the claustrum. In addition, ERalpha-ir cells were seen in the septum, basal forebrain, amygdala and hypothalamus. Double-labeled cells for ERalpha and calbindin-D(28k) were seen only in the ventrolateral part of the ventromedial hypothalamic nucleus. In contrast, the co-localization of ERalpha and parvalbumin or ChAT was not seen in any of the areas of the monkey forebrain examined. These observations suggest that estrogens, at least in part, via ERalpha regulate calbindin-D(28k) hypothalamic but not parvalbumin or ChAT containing neurons in select monkey forebrain regions.
Collapse
Affiliation(s)
- Sylvia Perez
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St., Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
90
|
Abstract
While it is undisputed that estrogens (1 beta-estradiol, E2) are mainly involved in skin physiology and operate as potent hair growth modulators, our knowledge about the estrogen target cells in skin and exact signaling pathways is still very limited. The current review provides an overview of estrogen effects on hair follicle cycling, cutaneous expression of estrogen receptors, and potential functions of estrogens in hair biology. We discuss potential target genes of estrogen receptor-mediated signaling in the skin, explore the interplay of estrogens with other hormones, growth factors and enzymes, and define major open questions in this intriguing and far too long neglected area of hair research.
Collapse
Affiliation(s)
- Franziska Conrad
- Department of Dermatology, University Hospital Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | | |
Collapse
|
91
|
Mufson EJ, Ginsberg SD, Ikonomovic MD, DeKosky ST. Human cholinergic basal forebrain: chemoanatomy and neurologic dysfunction. J Chem Neuroanat 2003; 26:233-42. [PMID: 14729126 DOI: 10.1016/s0891-0618(03)00068-1] [Citation(s) in RCA: 208] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The human cholinergic basal forebrain (CBF) is comprised of magnocellular hyperchromic neurons within the septal/diagonal band complex and nucleus basalis (NB) of Meynert. CBF neurons provide the major cholinergic innervation to the hippocampus, amygdala and neocortex. They play a role in cognition and attentional behaviors, and are dysfunctional in Alzheimer's disease (AD). The human CBF displays a continuum of large cells that contain various cholinergic markers, nerve growth factor (NGF) and its cognate receptors, calbindin, glutamate receptors, and the estrogen receptors, ERalpha and ERbeta. Admixed with these cholinergic neuronal phenotypes are smaller interneurons containing the m2 muscarinic acetylcholine receptor (mAChRs), NADPH-diaphorase, GABA, calcium binding proteins and several inhibitory neuropeptides including galanin (GAL), which is over expressed in AD. Studies using human autopsy material indicate an age-related dissociation of calbindin and the glutamate receptor GluR2 within CBF neurons, suggesting that these molecules act synergistically to induce excitotoxic cell death during aging, and possibly during AD. Choline acetyltrasnferease (ChAT) activity and CBF neuron number is preserved in the cholinergic basocortical system and up regulated in the septohippocampal system during prodromal as compared with end stage AD. In contrast, the number of CBF neurons containing NGF receptors is reduced early in the disease process suggesting a phenotypic silence and not a frank loss of neurons. In end stage AD, there is a selective reduction in trkA mRNA but not p75(NTR) in single CBF cells suggesting a neurotrophic defect throughout the progression of AD. These observations indicate the complexity of the chemoanatomy of the human CBF and suggest that multiple factors play different roles in its dysfunction in aging and AD.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Neurological Sciences and Alzheimer's Disease Center, Rush Presbyterian-St. Luke's Medical Center, Tech 2000, 2242 West Harrison St., Suite 200, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
92
|
Isgor C, Cecchi M, Kabbaj M, Akil H, Watson SJ. Estrogen receptor β in the paraventricular nucleus of hypothalamus regulates the neuroendocrine response to stress and is regulated by corticosterone. Neuroscience 2003; 121:837-45. [PMID: 14580933 DOI: 10.1016/s0306-4522(03)00561-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The function of the second nuclear estrogen receptor, estrogen receptor beta (ERbeta), in the brain is largely unknown. The present study tested whether 1) ERbeta in the paraventricular nucleus (PVN) of the hypothalamus has a direct role in the hypothalamic-pituitary-adrenal (HPA) axis-mediated stress function, and 2) whether corticosterone (CORT) can regulate ERbeta gene expression in the PVN in the intact, cycling female rat. To test the first hypothesis a pure estrogen receptor antagonist, ICI182, 780, was microinjected into the PVN bilaterally and stress-induced CORT response to an acute stressor (15 min restraint) was measured at 0, 15, 30, 60 and 90 min time points. Estrogen antagonist-injected rats showed inhibited CORT levels at the peak (15 min) of the stress response compared with vehicle-injected animals. To test the second hypothesis, ERbeta mRNA levels were measured in the PVN using in situ hybridization histochemistry following sham surgery, adrenalectomy, and adrenalectomy with low or high CORT replacement. Adrenalectomy reduced ERbeta mRNA expression in the PVN, whereas CORT replacement fully reversed this effect in a dose-dependent fashion. Both antagonist inhibition of CORT response and CORT-mediated regulation of ERbeta mRNA were found to be estrus cycle-dependent in the intact, cycling female. These data suggest that ERbeta in the PVN may critically modulate the HPA axis response to stress and is, in turn, regulated by circulating CORT.
Collapse
Affiliation(s)
- C Isgor
- Mental Health Research Institute, The University of Michigan School of Medicine, 205 Zina Pitcher Place, Ann Arbor, MI 48109-0720, USA.
| | | | | | | | | |
Collapse
|
93
|
Ikeda Y, Nagai A, Ikeda MA, Hayashi S. Sexually dimorphic and estrogen-dependent expression of estrogen receptor beta in the ventromedial hypothalamus during rat postnatal development. Endocrinology 2003; 144:5098-104. [PMID: 12960049 DOI: 10.1210/en.2003-0267] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ventromedial hypothalamus (VMH) is a sexually dimorphic region of the brain related to female reproductive behavior. The effect of estrogen in the adult rat VMH is thought to be mediated predominantly via estrogen receptor (ER)alpha, because this receptor is expressed at considerably higher levels than ER beta. The present study revealed, using in situ hybridization and immunohistochemistry, that both ER beta mRNA and protein were expressed in the ventrolateral portion of the caudal VMH, at remarkably higher levels during early postnatal development than in adulthood. In addition, the expression was sexually dimorphic, with females having significantly more ER beta-immunoreactive (-ir) cells than males, between postnatal d 5 (P5) and P14, although the sex difference was not significant by P21. Double-label immunofluorescence revealed that 66% of ER beta-ir cells coexpressed ER alpha in the caudal VMH of the P5 female rat. Furthermore, neonatal treatment with E2 benzoate down-regulated ER beta mRNA in the female rat VMH at P5 and decreased VMH ER beta-ir cells during the period between P5 and P14. In contrast to females, no differences in expression of ER beta mRNA or protein were detected between control and E2 benzoate-treated males. These results suggest that estrogen is involved in regulating the sexually dimorphic expression of ER beta in the VMH during early postnatal development of the rat.
Collapse
Affiliation(s)
- Yayoi Ikeda
- Laboratory of Endocrinology, Graduate School of Integrated Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan.
| | | | | | | |
Collapse
|
94
|
Pérez SE, Chen EY, Mufson EJ. Distribution of estrogen receptor alpha and beta immunoreactive profiles in the postnatal rat brain. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 145:117-39. [PMID: 14519499 DOI: 10.1016/s0165-3806(03)00223-2] [Citation(s) in RCA: 173] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The present study was conducted to identify the localization and possible contribution of the two estrogen receptor (ER) subtypes in the rat brain at postnatal (P) days 3, 7 and 14. Evaluation of the distribution of ERalpha and ERbeta immunoreactive (ir) nuclei did not reveal gender differences at the developmental point times examined. With the exception of the cerebral cortex, the pattern of staining for these ERs was unchanged across the postnatal ages examined. The distribution of ERalpha-ir nuclei was wider than ERbeta-ir during brain development. From P3, ERbeta and ERalpha-ir nuclei were found in different regions of the cerebral cortex, basal forebrain, amygdala, thalamus, hypothalamus, mesencephalon, pons, cerebellum and medulla oblongata. In addition, ERalpha-ir nuclei were exclusively detected in the hippocampal subfields, epithalamus and in several circumventricular organs. ERalpha and ERbeta dual immunofluorescence revealed positive nuclei in the medial part of the bed nucleus of the stria terminalis, periventricular preoptic nucleus and in caudal aspects of the ventrolateral part of the ventromedial hypothalamic nucleus. Although the functional significance of the dual expression of both ERs within the same nuclei remains unknown, it is possible that ERs play different roles in gene regulation within the same cell. The presence of ERs in diverse brain regions through early postnatal periods supports a potential role for estrogens in neural differentiation.
Collapse
Affiliation(s)
- Sylvia E Pérez
- Neurological Sciences, Rush Presbyterian St. Luke's Medical Center, Chicago, IL 60612, USA
| | | | | |
Collapse
|
95
|
Yamada K, Nozawa-Inoue K, Kawano Y, Kohno S, Amizuka N, Iwanaga T, Maeda T. Expression of estrogen receptor alpha (ER alpha) in the rat temporomandibular joint. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 274:934-41. [PMID: 12973717 DOI: 10.1002/ar.a.10107] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Numerous epidemiological studies have pointed out a higher frequency of temporomandibular disorder (TMD) in women than in men, which indicates the involvement of a sex hormone, such as estrogen, in the pathogenesis of TMD. Although estrogen is known to play pivotal roles in osteoarthrosis or rheumatoid arthritis in systemic joints, there have been few reports about the role of estrogen in the temporomandibular joint (TMJ). The effect of estrogen is generally mediated by the estrogen receptors (ERs) ER alpha (the predominant type) and ER beta. In this study we examined the expression of ER alpha protein and mRNA in the TMJ of adult male rats by immunocytochemistry and in situ hybridization histochemistry. Intense ER alpha immunoreactivity was localized in the synovial lining cells, stromal cells in the articular disc, and chondrocytes in the TMJ. These ER alpha-immunopositive synovial lining cells are characteristic of cytoplasmic processes identified with confocal and immunoelectron microscopy, which indicates that they are synovial type B cells. In situ hybridization histochemistry confirmed intense signals for ER alpha in the synovial lining cells and the sublining fibroblasts at mRNA levels. The nuclei of chondrocytes showed an intense immunoreaction for ER alpha in the maturative and hypertrophic layers of the articular cartilage. In addition to the nuclear localization of ER alpha, a weak immunoreaction appeared in the cytoplasm of some ER alpha-positive cells. These findings support the hypothesis that TMJ tissue-at least in the male rat-has the potential to be an estrogen target tissue.
Collapse
Affiliation(s)
- Kazuho Yamada
- Division of Removable Prosthodontics, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | | | | | | | | |
Collapse
|
96
|
Flores CA, Shughrue P, Petersen SL, Mokha SS. Sex-related differences in the distribution of opioid receptor-like 1 receptor mRNA and colocalization with estrogen receptor mRNA in neurons of the spinal trigeminal nucleus caudalis in the rat. Neuroscience 2003; 118:769-78. [PMID: 12710984 DOI: 10.1016/s0306-4522(02)01000-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We recently reported that exogenously applied orphanin FQ, the endogenous ligand for opioid receptor-like 1 (ORL(1)) receptor, produces sex-specific modulation of trigeminal nociception, and that estrogen contributes to these sex-related differences. Estrogen could produce these sex-related differences by altering the expression of the ORL(1)-receptor gene in the trigeminal nucleus caudalis. Utilizing in situ hybridization, we compared levels of ORL(1) receptor mRNA and investigated its colocalization with estrogen receptor mRNA in trigeminal neurons. Our results showed that in male rats, ORL(1) receptor mRNA is abundantly expressed in the rostral part of the trigeminal nucleus caudalis, and at the junction of caudalis and interpolaris (Vc/Vi). In comparison with males, levels of ORL(1) receptor mRNA were not significantly different in proestrus females, but were significantly higher in the rostral trigeminal nucleus caudalis and at the junction of Vc/Vi of diestrus females. In addition, ovariectomy raised the levels in the rostral trigeminal nucleus caudalis, and at the junction of Vc/Vi. Levels were reduced to proestrus levels in these regions following estradiol replacement. Our results also showed that ORL(1) receptor mRNA is present in majority of estrogen receptor (alpha and/or beta) mRNA-containing neurons. We conclude that there are sex-related differences in the ORL(1)-receptor gene expression in the trigeminal nucleus caudalis, which appear to be determined in part by estrogen levels.
Collapse
Affiliation(s)
- C A Flores
- Department of Anatomy and Physiology, Meharry Medical College, 1005 D. B. Todd Boulevard, Nashville, TN 37208, USA
| | | | | | | |
Collapse
|
97
|
Mowa CN, Usip S, Collins J, Storey-Workley M, Hargreaves KM, Papka RE. The effects of pregnancy and estrogen on the expression of calcitonin gene-related peptide (CGRP) in the uterine cervix, dorsal root ganglia and spinal cord. Peptides 2003; 24:1163-74. [PMID: 14612187 DOI: 10.1016/j.peptides.2003.07.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Before parturition the uterine cervix undergoes a ripening process ("softens" and dilates) to allow passage of the fetus at term. The exact mechanism(s) responsible for cervical ripening are unknown, though a role for peptidergic sensory neurons is emerging. Previous work demonstrated that administration of substance P (SP) to ovariectomized rats caused events associated with cervical ripening, that production of SP in cervix-related dorsal root ganglion (DRG) is estrogen responsive, and that release of SP from neurons terminating in the cervix and spinal cord peaks prior to parturition. The present study was designed to test the hypothesis that calcitonin gene-related peptide (CGRP), a neuropeptide co-stored with SP in many sensory neurons, undergoes changes with pregnancy and hormonal environment. Immunohistochemistry, in situ hybridization, reverse transcriptase-polymerase chain reaction (RT-PCR) and radioimmunoassay (RIA) were used to investigate CGRP in L6-S1 DRG, spinal cord and cervix during pregnancy and the role of estrogen in CGRP synthesis. CGRP-immunoreactive primary sensory neurons expressed estrogen receptors (ER-alpha and ER-beta). In the cervix, CGRP concentrations decreased, but in the L6-S1 DRG and the spinal cord segments, CGRP levels increased, with peak effects observed at day 20 of gestation. CGRP mRNA synthesis increased in DRG over pregnancy. Sensory neurons of ovariectomized rats treated with estrogen showed increased CGRP mRNA synthesis in a dose-related manner, an effect blocked by the ER antagonist ICI 182 780. From these results, we postulate that synthesis of CGRP in L6-S1 DRG and utilization in the cervix increase over pregnancy and this synthesis is the under influence of the estrogen-ER system. Collectively, these data are consistent with the hypothesis that CGRP plays a role in cervical ripening and, consequently in the birth process.
Collapse
Affiliation(s)
- C N Mowa
- Department of Neurobiology, Northeastern Ohio Universities, College of Medicine, P.O. Box 95, 4209 State Rt. 44, Rootstown, OH 44272 , USA
| | | | | | | | | | | |
Collapse
|
98
|
Schausi D, Tiffoche C, Thieulant ML. Regulation of the intronic promoter of rat estrogen receptor alpha gene, responsible for truncated estrogen receptor product-1 expression. Endocrinology 2003; 144:2845-55. [PMID: 12810539 DOI: 10.1210/en.2003-0024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have characterized the intronic promoter of the rat estrogen receptor (ER) alpha gene, responsible for the lactotrope-specific truncated ER product (TERP)-1 isoform expression. Transcriptional regulation was investigated by transient transfections using 5'-deletion constructs. TERP promoter constructs were highly active in MMQ cells, a pure lactotrope cell line, whereas a low basal activity was detected in alphaT3-1 gonadotrope cells or in COS-7 monkey kidney cells. Serial deletion analysis revealed that 1) a minimal -693-bp region encompassing the TATA box is sufficient to allow lactotrope-specific expression; 2) the promoter contains strong positive cis-acting elements both in the distal and proximal regions, and 3) the region spanning the -1698/-1194 region includes repressor elements. Transient transfection studies, EMSAs, and gel shifts demonstrated that estrogen activates the TERP promoter via an estrogen-responsive element (ERE1) located within the proximal region. Mutation of ERE1 site completely abolishes the estradiol-dependent transcription, indicating that ERE1 site is sufficient to confer estrogen responsiveness to TERP promoter. In addition, ERalpha action was synergized by transfection of the pituitary-specific factor Pit-1. EMSAs showed that a single Pit-1 DNA binding element in the vicinity of the TATA box is sufficient to confer response by the TERP promoter. In conclusion, we demonstrated, for the first time, that TERP promoter regulation involves ERE and Pit-1 cis-elements and corresponding trans-acting factors, which could play a role in the physiological changes that occur in TERP-1 transcription in lactotrope cells.
Collapse
Affiliation(s)
- Diane Schausi
- Université de Rennes I, Interactions Cellulaires et Moléculaires, Equipe Information et Programmation Cellulaires, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 6026, Campus de Beaulieu, 35042 Rennes Cedex, France
| | | | | |
Collapse
|
99
|
Kametaka M, Kume A, Okada T, Mizukami H, Hanazono Y, Ozawa K. Reduction of CTLL-2 cytotoxicity by induction of apoptosis with a Fas-estrogen receptor chimera. Cancer Sci 2003; 94:639-43. [PMID: 12841875 PMCID: PMC11160217 DOI: 10.1111/j.1349-7006.2003.tb01496.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2003] [Accepted: 04/30/2003] [Indexed: 11/30/2022] Open
Abstract
Allogeneic bone marrow transplantation and donor lymphocyte infusion are powerful treatments for chemotherapy-resistant leukemia. Tumor eradication is attributed to a graft-versus-leukemia reaction by the donor-derived cytotoxic T lymphocytes (CTLs), but the same cell population may cause severe graft-versus-host disease. One strategy to suppress harmful CTL activity is to incorporate a suicide gene into the donor lymphocytes prior to infusion, and to destroy these cells when they aggressively attack nonmalignant host tissues. In this study, we investigated the feasibility of using a Fas-estrogen receptor fusion protein (MfasER) to control T cell-mediated cytotoxicity, based on our previous finding that the chimera transmits a Fas-mediated death signal through activation by estrogen binding. A murine CTL line CTLL-2 was transfected with a vector encoding MfasER, and the growth, viability and cytotoxic activity of the transfected cells (CTLL/MfasER) were analyzed. The expression of apoptosis-related proteins such as Fas ligand and perforin was also investigated. In the absence of estrogen, CTLL/MfasER showed similar growth to parental CTLL-2, and the killing activity was preserved. Addition of 10 (-7) M estrogen induced a rapid apoptosis of CTLL/MfasER, and the cytotoxicity was severely impaired. A decrease of Fas ligand and perforin in the estrogen-treated CTLL/MfasER was seen in an immunoblot analysis. These functional and biochemical analyses showed that the estrogen-inducible apoptosis in MfasER-expressing CTLs rapidly terminated their target cell killing. The feasibility of using the MfasER-estrogen system to control graft-versus-host disease was demonstrated.
Collapse
Affiliation(s)
- Minako Kametaka
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Tochigi 329-0498, Japan
| | | | | | | | | | | |
Collapse
|
100
|
Choi CY, Habibi HR. Molecular cloning of estrogen receptor alpha and expression pattern of estrogen receptor subtypes in male and female goldfish. Mol Cell Endocrinol 2003; 204:169-77. [PMID: 12850291 DOI: 10.1016/s0303-7207(02)00182-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Estrogen plays an important role in the regulation of gonadotropin production in vertebrates. In this study, we isolated the estrogen receptor (ER) alpha cDNA from the goldfish pituitary. Primers for ERalpha were designed based on the similarity of selected regions (C and E domains) of known ER genes. Full-length cDNA sequence for ERalpha was determined by 3' and 5' RACE procedures. ERalpha cDNA clone was found to contain 2087 nucleotides including an open reading frame that encodes 564 amino acids, with a molecular weight of 62.8 kDa. We also cloned ERbeta-1 and ERbeta-2 from the published information and investigated the expression pattern of these ER subtypes in a variety of tissues in male and female goldfish by reverse transcriptase-polymerase chain reaction (RT-PCR). Significant variations in the relative expression of ERalpha, ERbeta-1 and ERbeta-2 were observed in different tissues in male and female goldfish. Pituitary was found to have the highest expression level of ERalpha in both male and female goldfish. Significantly, lower levels of ERalpha expression were observed in the brain, ovary, testis, liver, muscle, heart and intestine. Ovary and testis were found to have higher transcript levels of ERbeta-1 with much lower levels in the brain, pituitary, liver, muscle and heart. The ERbeta-2 was found to be expressed strongly in the pituitary followed by intestine with lower expression in other tissues. The present study provides molecular characterization of ERalpha, and information on tissue specific distribution of different ER subtypes in male and female goldfish.
Collapse
Affiliation(s)
- C Y Choi
- Department of Biological Sciences, The University of Calgary, 2500 University Drive NW, Calgary, AB, Canada T2N 1N4
| | | |
Collapse
|