Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Exp Physiol ; 95(7): 819-28, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20360425

ABSTRACT

The ciliary beat frequency (CBF) of guinea-pig fimbria during the ovarian cycle was measured by video microscopy using a high-speed camera (500 Hz). In the follicular phase, with increasing concentrations of beta-oestradiol ([betaE(2)]) and a low concentration of progesterone ([PRG]), CBF increased from 13.5 to 16 Hz. In the ovulatory phase, with further increase of [betaE(2)], CBF decreased gradually from 16 to 13.5 Hz. In the early luteal phase, with low [PRG] and [betaE(2)], CBF increased to 17 Hz; however, in the middle luteal phase, with increasing [PRG], CBF decreased (12 Hz), and in the late luteal phase, with decreasing [PRG], CBF increased to 15 Hz. Then, in the resting phase, with low [betaE(2)] and [PRG], CBF decreased immediately to 14 Hz. The CBF of the fimbria was measured in guinea-pigs treated with beta-oestradiol benzoate (betaE(2)B) or medroxyprogesterone (mPRG). A low dose of betaE(2)B increased CBF to 14.5 Hz, whereas a high dose decreased it to 11 Hz. A betaE(2) receptor blocker, ICI-182,780, abolished the betaE(2)B-induced CBF changes and maintained CBF at 12.0 Hz. Medroxyprogesterone decreased CBF to 12.5 Hz, and mifepristone (a PRG receptor blocker) abolished the mPRG-induced CBF decrease and maintained CBF at 15 Hz. The addition of both blockers increased CBF to 18 Hz, suggesting that activation of betaE(2) or PRG receptors decreases the CBF of the fimbria. In conclusion, a moderate [betaE(2)] increase maintains a high CBF (15.5 Hz) in the follicular phase, and then further [betaE(2)] increase decreases CBF to 13.5 Hz in the ovulatory phase. In the early and late luteal phase, low [betaE(2)] and [PRG] increase CBF to 17 and 15 Hz, respectively, and in the middle luteal phase a high [PRG] decreases CBF (to 12 Hz). Thus, the CBF of the fimbria was controlled by signals via betaE(2) and PRG receptors in guinea-pigs.


Subject(s)
Cilia/drug effects , Cilia/physiology , Estradiol/pharmacology , Estrous Cycle/physiology , Progesterone/pharmacology , Animals , Estradiol/analogs & derivatives , Estrous Cycle/drug effects , Fallopian Tubes/physiology , Female , Fulvestrant , Guinea Pigs , Medroxyprogesterone/pharmacology , Microscopy, Video , Mifepristone/pharmacology , Ovulation/physiology , Receptors, Estradiol/physiology , Receptors, Progesterone/physiology
2.
Science ; 232(4757): 1540-3, 1986 Jun 20.
Article in English | MEDLINE | ID: mdl-3715461

ABSTRACT

The hormone 17 beta-estradiol acts through its receptor system to induce MCF-7 human breast cancer cells to form tumors in athymic mice. In vitro studies have identified the production of estrogen-induced growth factors from MCF-7 cells that may have a role in growth control. These induced growth factors were sufficient to stimulate MCF-7 tumor growth in ovariectomized athymic mice, thus partially replacing estradiol. Growth factors may act as estrogen-induced "second messengers" in estrogen-responsive growth of human breast cancer.


Subject(s)
Breast Neoplasms/pathology , Estradiol/physiology , Receptors, Estradiol/physiology , Receptors, Estrogen/physiology , Animals , Cell Division , Cell Line , Culture Media , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovariectomy , Transplantation, Heterologous
3.
Gen Comp Endocrinol ; 161(1): 58-61, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18952087

ABSTRACT

The orphan G protein coupled receptor, GPR30, has the characteristics of a high affinity, specific estrogen membrane receptor on Atlantic croaker oocytes and mediates estrogen inhibition of oocyte maturation in this perciform fish. In order to determine the broad applicability of these findings to other teleosts, similar experiments were conducted in a cyprinid fish, zebrafish, in the present study. GPR30 mRNA expression was detected in zebrafish oocytes but not in the ovarian follicular cells. Both spontaneous and 17, 20beta-dihyroxy-4-pregnen-3-one (DHP)-induced maturation of follicle-enclosed zebrafish oocytes was significantly decreased when they were incubated with either estradiol-17beta, or the GPR30 agonists, ICI 182 780 and tamoxifen, or with the GPR30 specific agonist G-1. On the other hand spontaneous oocyte maturation increased two-fold when zebrafish ovarian follicles were incubated with an aromatase inhibitor, ATD. Moreover, the stimulatory effects of ATD on germinal vesicle breakdown (GVBD) were partially reversed by co-treatment with 100 nM of E2 or G-1. These results suggest that endogenous estrogens acting through GPR30 are involved in maintaining meiotic arrest of zebrafish oocytes.


Subject(s)
Estradiol/metabolism , Oocytes/physiology , Receptors, Estradiol/physiology , Receptors, G-Protein-Coupled/physiology , Zebrafish Proteins/physiology , Androstatrienes/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Cyclopentanes/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Fulvestrant , Hydroxyprogesterones/pharmacology , Quinolines/pharmacology , Receptors, Estradiol/agonists , Receptors, G-Protein-Coupled/agonists , Tamoxifen/pharmacology , Zebrafish
4.
Endocrinology ; 147(9): 4056-66, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16809439

ABSTRACT

There is an urgent need to identify and develop a new generation of therapeutic agents and systemic therapies targeting the estradiol (E2)/estrogen receptor (ER) signaling in breast cancer. In this regard, new information on the mechanisms of E2/ER function and/or cross talk with other prosurvival cascades should provide the basis for the development of other ideal anti-E2 therapies with the intent to enhance clinical efficacy, reduce side effects or both. Our very recent assessment of the mechanisms by which cancer-associated increased lipogenesis and its inhibition alters the E2/ER signaling discovered that fatty acid synthase (FASN), the enzyme catalyzing the terminal steps in the de novo biosynthesis of long-chain fatty acids, differentially modulates the state of sensitivity of breast and endometrial cancer cells to E2-stimulated ER transcriptional activation and E2-dependent cell growth and survival: 1) pharmacological inhibition of FASN activity induced a dramatic augmentation of E2-stimulated ER-driven gene transcription, whereas interference (RNAi)-mediated silencing of FAS gene expression drastically lowered E2 requirements for optimal activation of ER transcriptional activation in breast cancer cells; conversely, pharmacological and RNAi-induced inhibition of FASN worked as an antagonist of E2- and tamoxifen-dependent ER transcriptional activity in endometrial adenocarcinoma cells; 2) pharmacological and RNAi-induced inhibition of FASN synergistically enhanced E2-mediated down-regulation of ER protein and mRNA expression in breast cancer cells, whereas specific FASN blockade resulted in a marked down-regulation of E2-stimulated ER expression in endometrial cancer cells; and 3) FASN inhibition decreased cell proliferation and cell viability by promoting apoptosis in hormone-dependent breast and endometrial cancer cells. In this review we propose that, through a complex mechanism involving the regulation of MAPK/ER cross talk as well as critical E2-related proteins including the Her-2/neu (erbB-2) oncogene and the cyclin-dependent kinase inhibitors p21(WAF1/CIP1) and p27(Kip1), a previously unrevealed connection exists between FASN and the genomic and nongenomic ER activities in breast and endometrial cancer cells. From a clinical perspective, we suggest that if chemically stable FASN inhibitors or cell-selective systems able to deliver RNAi targeting FASN gene demonstrate systemic anticancer effects of FASN inhibition in vivo, additional preclinical studies to characterize their anti-breast cancer actions should be of great interest as the specific blockade of FASN activity may also provide a protective means against endometrial carcinoma associated with tamoxifen-based breast cancer therapy.


Subject(s)
Breast Neoplasms/drug therapy , Endometrial Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Fatty Acid Synthases/antagonists & inhibitors , Selective Estrogen Receptor Modulators/therapeutic use , Estradiol/physiology , Fatty Acid Synthases/genetics , Fatty Acid Synthases/physiology , Fatty Acids/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , RNA, Small Interfering/pharmacology , Receptors, Estradiol/physiology , Signal Transduction
5.
Cancer Res ; 52(18): 5082-8, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1516065

ABSTRACT

Epidermal growth factor (EGF) receptor is inversely related to expression of estrogen receptor (ER) and progesterone receptor in primary breast tumors and is a negative predictor for response to endocrine therapy. To investigate a possible causal role of EGF receptor expression in breast cancer progression to hormone independence, we have created an experimental cell system. Epidermal growth factor receptor complementary DNA was introduced in estrogen-dependent ZR-75-1 breast cancer cells, and the resulting ZR/HERc cells exhibited a mitogenic response to epidermal growth factor, thus bypassing estrogen dependence. This EGF-induced proliferation could not be inhibited by antiestrogens. In addition, we noted changes in cell morphology and keratin expression of EGF-stimulated ZR/HERc cells, suggestive of an altered differentiation state. Furthermore, intolerance of functional ER and EGF receptor signal transduction pathways in ZR/HERc cells was observed during simultaneous activation, which possibly explains the inverse relationship of ER and EGF receptor expression in primary tumors. In contrast to the parental cells, ZR/HERc cells rapidly progressed to a stable ER-negative phenotype when cultured in the presence of the antiestrogen hydroxy-tamoxifen. These results suggest a possible role for EGF receptor in progression of breast cancer to hormone independence.


Subject(s)
Breast Neoplasms/pathology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Estrogens/physiology , Cell Differentiation/drug effects , Cell Division/drug effects , Drug Resistance , ErbB Receptors/genetics , Gene Expression , In Vitro Techniques , Receptors, Estradiol/physiology , Signal Transduction , Tamoxifen/pharmacology , Transfection , Tumor Cells, Cultured
6.
Oncogene ; 12(4): 753-63, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8632897

ABSTRACT

HMG-CoA reductase inhibitors, such as Lovastatin and Simvastatin, cause cell cycle arrest by interfering with the mitogenic activity of mitogens present in culture media. Cells are induced to pause in G1 and can readily resume growth upon removal of the enzymatic block. Estrogens, acting via their nuclear receptor, are mitogens for different normal and transformed cell types, where they foster cell cycle progression and cell division. In estrogen-responsive MCF-7 human breast cancer cells, but not in non responsive cells, 17 beta-estradiol (E2) induces cells arrested with Lovastatin or Simvastatin to proliferate in the presence of inhibitor, without restoring HMG-CoA reductase activity or affecting the protein prenylation pattern. Mitogenic stimulation of G1-arrested MCF-7 cells with E2 includes primary transcriptional activation of c-fos, accompanied by transient binding in vivo of the estrogen receptor and/or other factors to the ERE and the estrogen-responsive DNA region of this proto-oncogene, as detected by dimethylsulphate genomic footprinting analysis. Mitogenic stimulation of growth-arrested MCF-7 cells by E2 occurs, under these conditions, without evident activation of ERK-1 and -2 kinases, and thus independently from the mitogen-responsive signal transduction pathways that converge on these enzymes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle/drug effects , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-fos/metabolism , Base Sequence , Breast Neoplasms , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cholesterol/biosynthesis , Enzyme Activation , Female , G1 Phase/drug effects , Genes, fos/drug effects , Humans , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Oligodeoxyribonucleotides , Proto-Oncogene Mas , Receptors, Estradiol/physiology , Simvastatin , Transcriptional Activation/drug effects , Tumor Cells, Cultured
7.
Mol Endocrinol ; 10(3): 217-29, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8833651

ABSTRACT

The orphan nuclear receptor steroidogenic factor-1 (SF-1) regulates the expression of several genes involved in the reproductive function and development of the adrenal, the gonads, and the pituitary gonadotropes. It also confers the gonadotrope-specific expression of the glycoprotein hormone a subunit gene by the binding to a gonadotrope-specific element (GSE). In this study, we have shown that SF-1 transactivates the salmon gonadotropin II beta subunit (sGTHII beta) gene expression. SF-1 alone offered a slight but significant enhancement on sGTHII beta promoter activity (7.2 +/- 0.6 fold). However, it stimulated sGTHII beta gene expression dramatically (127 +/- 37 fold) when combined with the estrogen receptor (ER). This synergistic interaction was specific for sGTHII beta promoter as well as for both SF-1 and ER and was estradiol-dose dependent. 5'-Deletion studies of the sGTHII beta promoter identified two putative SF-1 binding sites (GSE) and one previously identified proximal estrogen-responsive element (pERE) at -274 bp involved in this activation. The two GSE sequences located at -354 bp (sGSE(3) and -162 bp (sGSE(2) upstream of the transcription site, although imperfect as compared with the consensus GSE, bound specifically to the in vitro-translated mouse SF-1 protein. 5'-Deletion studies, competition experiments, and site-directed mutagenesis showed that binding to pERE and GSE(2) were necessary for the SF-1/ER synergistic effect. These studies suggest that the synergistic interaction of SF-1 and ER, possibly through cooperative binding or protein-protein interaction, is essential in conferring a cell type-specific expression of the GTHII beta subunit gene.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Gonadotropins, Pituitary/genetics , Receptors, Estradiol/physiology , Salmon/genetics , Transcription Factors/physiology , Animals , Consensus Sequence , Fushi Tarazu Transcription Factors , Genes , Gonadotropins, Pituitary/biosynthesis , HeLa Cells , Homeodomain Proteins , Humans , Mice , Mutagenesis, Site-Directed , Organ Specificity , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Steroidogenic Factor 1 , Stimulation, Chemical , Transcription, Genetic , Transfection
9.
Endokrynol Pol ; 56(3): 322-6, 2005.
Article in Polish | MEDLINE | ID: mdl-16350726

ABSTRACT

Classical action of steroid hormones, called genomic, includes binding to their intracellular receptor, require hours or days to occur and require transcriptional effects with subsequent modulation of protein expression. Some of the biological effects induced by steroids, and mainly by sex steroids, take place within seconds or few minutes, time far too fast to be due to the genomic changes. The rapid, nongenomic action of estradiol are attributed to membrane action, probably through variety of proteins present in cell membrane. The rapid effects of steroid hormones are manifold, ranging from activation of protein and tyrosine kinases, G proteins, and modulation of ion channels. The nongenomic way of action includes also non-direct control of processes of transcription and gene expression. There are at least three different way to interact with cell membrane. Steroids may change membrane fluidity, without binding to any known protein or receptor. Another way is allosteric modulation of non-specific for steroid hormones receptors, or structural and enzymatic protein present in cell membrane. Evidence suggests that the classical steroid receptors can be localized at the plasma membrane, triggering signals typical for G-proteins coupled receptors. Physiological significance of nongenomic action of steroids needs to be elucidated.


Subject(s)
Receptors, Cell Surface/physiology , Receptors, Estradiol/physiology , Animals , Binding Sites , Cell Membrane/metabolism , Estradiol , Humans , Receptors, G-Protein-Coupled/physiology
10.
Endocrinology ; 116(1): 148-53, 1985 Jan.
Article in English | MEDLINE | ID: mdl-3964746

ABSTRACT

The nuclear binding kinetics of uterine 17 beta-estradiol (E2) receptors (UER) were studied throughout aging in intact and castrated (OVX) mice. When compared to young animals, 15- to 18-month-old mice showed a significant reduction in their total cytosolic (0.526 vs. 0.405 pmol/uterus; P less than 0.05) and nuclear (0.37 vs. 0.16 pmol/uterus; P less than 0.01) UER content, whereas the affinity (Ka) for estrogens remained constant (0.8-1.6 X 10(9) M-1). This age-related decrease in UER was preceded by a blunted and retarded nuclear binding of UER at 10-14 months of age, which was further accentuated after transition from perimenopause. Ovariectomy (OVX), whether performed neonatally or in adulthood, reduced the total concentration of cytosolic and nuclear UER in each age group studied, but did not prevent this reduced nuclear binding observed in middleaged mice. However, when standardized per tissue protein, the mean number of cytosolic UER from young and middle-aged, but not old, mice was reduced by 50% after neonatal OVX (176.5, 178.4, and 218.8 fmol/mg protein, respectively), whereas it remained unchanged when OVX was performed in adulthood and the animals subsequently studied at peri- and postmenopausal ages (326.3 and 283.3 fmol/mg protein, respectively). Daily administration of a physiological dose of E2 for 7 days to OVX mice of each age group induced maximal synthesis of UER in young animals, but not in peri- and postmenopausal ones; in peri- and postmenopausal animals, this was paralleled by reduced uterotropic responses despite similar increments in plasma E2. These results suggest an age-related, gonad-independent decline in the number of functional UER early in reproductive aging.


Subject(s)
Aging , Uterus/metabolism , Animals , Castration , Cell Nucleus/metabolism , Cytosol/metabolism , Estradiol/pharmacology , Female , Kinetics , Menopause , Mice , Mice, Inbred C57BL , Receptors, Estradiol/drug effects , Receptors, Estradiol/physiology , Uterus/drug effects , Uterus/growth & development
11.
Endocrinology ; 142(4): 1669-77, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250949

ABSTRACT

Estradiol (E(2))-signaling is widely considered to be exclusively mediated through the transcription-regulating intracellular estrogen receptor (ER) alpha and ERbeta. The aim of this study was to investigate transcription-independent E(2)-signaling in mouse IC-21 macrophages. E(2) and E(2)-BSA induce a rapid rise in the intracellular free Ca(2+) concentration ([Ca(2+)](i)) of Fura-2 loaded IC-21 cells as examined by spectrofluorometry. These changes in [Ca(2+)](i) can be inhibited by pertussis toxin, but not by the ER-blockers tamoxifen and raloxifene. The E(2)-signaling initiated at the plasma membrane is mediated through neither ERalpha nor ERbeta, but rather through a novel G protein-coupled membrane E(2)-receptor as revealed by RT-PCR, flow cytometry, and confocal laser scanning microscopy. A special feature of this E(2)-receptor is its sequestration upon agonist stimulation. Sequestration depends on energy and temperature, and it proceeds through a clathrin- and caveolin-independent pathway.


Subject(s)
Estradiol/physiology , Receptors, Cell Surface/physiology , Receptors, Estradiol/physiology , Signal Transduction/physiology , Animals , Calcium/metabolism , Cells, Cultured , Estradiol/metabolism , Flow Cytometry , Fluorescein-5-isothiocyanate , Macrophages/metabolism , Mice , Microscopy, Confocal , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Serum Albumin, Bovine/metabolism
12.
Endocrinology ; 136(5): 2309-19, 1995 May.
Article in English | MEDLINE | ID: mdl-7536668

ABSTRACT

We report that growth of LNCaP human prostate cancer cells is significantly stimulated (up to 120% above control) by physiological estradiol (E2) concentrations. This growth increase appears to be comparable to that induced by either testosterone or dihydrotestosterone, as also reported by others. This paper presents novel illustrative evidence for estrogen-binding proteins and messenger RNA transcripts in LNCaP cells. In fact, 1) the reverse transcriptase-polymerase chain reaction system documented normal messenger RNA for estrogen receptors (ER); 2) the radioligand binding assay allowed the detection of high affinity, reduced capacity binding sites in both soluble and nuclear cell fractions; and 3) the immunocytochemical analysis showed a consistently intensive staining for both ER and progesterone receptors. Compared to other human estrogen-responsive mammary cancer cells, MCF7 and ZR75-1, ER expression in LNCaP cells was not significantly lower, as shown by levels of the ER transcripts, number of sites per cell, or femtomoles per mg DNA as well as the percentage and intensity of immunocytochemical staining. A relative estimate of ER expression obtained by matching LNCaP with another human prostate cancer cell line, PC3, always displayed significantly and consistently higher levels in LNCaP cells. The detection of relatively high type I ER content in either cell compartment of LNCaP cells was paralleled by a highly intensive staining for progesterone receptors. In addition, evidence that the synthetic androgen R1881 did not compete for type I binding of E2 and that any E2-induced growth was completely reversed by the pure antiestrogen ICI-182,780, but unaffected by the antiandrogen Casodex, clearly suggests that the biological response of LNCaP cells to E2 is mediated via its own receptor.


Subject(s)
Cell Division/physiology , Estradiol/pharmacology , Receptors, Estradiol/physiology , Androgen Antagonists/pharmacology , Base Sequence , Breast Neoplasms , Cell Division/drug effects , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , DNA Primers , DNA, Neoplasm/metabolism , Dihydrotestosterone/pharmacology , Female , Flutamide/analogs & derivatives , Flutamide/pharmacology , Gene Expression , Humans , Kinetics , Male , Molecular Sequence Data , Polymerase Chain Reaction , Prostate-Specific Antigen/analysis , Prostatic Neoplasms , Radioligand Assay , Receptors, Estradiol/biosynthesis , Receptors, Estradiol/drug effects , Transcription, Genetic , Tumor Cells, Cultured
13.
Endocrinology ; 135(4): 1353-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7925096

ABSTRACT

Estradiol increases the number of GnRH receptors in the ewe. Although results from studies conducted in vitro indicate that progesterone may have a negative influence on the number of ovine GnRH receptors, this effect of progesterone has not been documented in vivo. To explore the regulation of GnRH receptors at the level of gene expression, a partial complementary DNA (cDNA) encoding ovine GnRH receptor was isolated using reverse transcription and polymerase chain reaction methodology. This partial cDNA (701 basepairs) was used to isolate a full-length cDNA encoding GnRH receptor from an ovine pituitary cDNA library. Northern blot analysis of RNA from ovine pituitary glands using the partial cDNA as a molecular probe revealed four messenger RNA (mRNA) transcripts at 5.6, 3.8, 2.1, and 1.3 kilobases. In some samples, a fifth transcript at 0.8 kilobases was also evident. GnRH receptor mRNA was not detected in ovine brain, heart, kidney, adrenal, or liver tissues. To examine the regulation of GnRH receptor mRNA and GnRH receptors during the early preovulatory period, relationships among steady state concentrations of GnRH receptor mRNA, numbers of GnRH receptors, and circulating concentrations of progesterone and estradiol during luteolysis were characterized. We hypothesized that during luteolysis, decreased concentrations of progesterone would be associated with increased concentrations of GnRH receptor mRNA and increased numbers of GnRH receptors. On day 11 or 12 of the estrous cycle, luteolysis was induced in 14 ewes by treatment with prostaglandin F2 alpha (PGF2 alpha). Four ewes were treated with saline (saline controls). Anterior pituitary tissue was collected 4 h (n = 4), 12 h (n = 5), and 24 h (n = 5) after treatment with PGF2 alpha or 24 h after treatment with saline and from four untreated ewes on day 11 or 12 of the estrous cycle (untreated controls). Twelve hours after treatment with PGF2 alpha, circulating concentrations of progesterone had decreased (P < 0.05) to 46% of the control values; however, concentrations of estradiol were not different from those in control ewes. Concentrations of GnRH receptor mRNA increased 2-fold during luteolysis and were higher than control values 12 h after PGF2 alpha treatment (P < 0.05). This increase in GnRH receptor mRNA was not accompanied by an increase in the number of GnRH receptors. Twenty-four hours after treatment with PGF2 alpha, concentrations of progesterone in PGF2 alpha-treated ewes had decreased (P < 0.05) to 15% of control values, whereas concentrations of estradiol had increased (P < 0.05) to 321% of control values.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Follicular Phase/physiology , RNA, Messenger/genetics , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Sheep/physiology , Animals , Base Sequence , Blotting, Northern , DNA/analysis , DNA/genetics , Dinoprost/pharmacology , Dose-Response Relationship, Drug , Estradiol/blood , Estradiol/pharmacology , Female , Follicular Phase/blood , Gene Expression Regulation , Molecular Sequence Data , Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/ultrastructure , Polymerase Chain Reaction , Progesterone/blood , RNA, Messenger/analysis , Receptors, Estradiol/analysis , Receptors, Estradiol/physiology , Receptors, LHRH/analysis , Time Factors
14.
Bone ; 27(1): 41-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10865207

ABSTRACT

Although estrogen is known to induce new bone formation in the long bones of female mice, this response is only thought to occur following administration of high doses, suggesting that it may not be mediated by a conventional estrogen receptor. To address this question further, we first examined the stereospecificity of this response by comparing the potency of 17beta-estradiol (E(2)) in stimulating cancellous bone formation at the proximal tibial metaphysis of intact female mice with that of the relatively inactive stereoisomer, 17alpha-estradiol (alphaE(2)). We found that E(2) was significantly more potent than alphaE(2), as assessed by histomorphometry. To provide further evidence for an estrogen-receptor-mediated process, we examined whether E(2)-induced osteogenesis in intact female mice could be inhibited by the estrogen receptor antagonist, ICI 182,780 (ICI). Although ICI itself had no effect on histomorphometric indices of the proximal tibial metaphysis when given alone, it significantly inhibited the osteogenic response to E(2). Finally, we examined the dose dependency of E(2)-induced osteogenesis at the proximal tibial metaphysis in intact mice. We found that E(2) stimulated cancellous bone formation in a dose-dependent manner over a wide dose range (i. e., 1-4000 microg/kg per day), with significant increases observed at doses of 4 microg/kg per day and beyond. Our results raise the possibility that estrogen-induced osteogenesis in the mouse represents an estrogen-receptor-mediated response that is not confined solely to supraphysiological estrogen levels.


Subject(s)
Estradiol/pharmacology , Osteogenesis/drug effects , Osteogenesis/physiology , Receptors, Estradiol/physiology , Animals , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred CBA
15.
J Mol Endocrinol ; 19(3): 249-57, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9460646

ABSTRACT

In the present study, the induction of the phosphoinositide signal transduction pathway by 17 beta-oestradiol has been demonstrated in rat vaginal epithelial cells (VEC). We have shown an increase in the metabolism of phosphoinositol lipids by 3H-myoinositol incorporation as well as production of inositol phosphate in VEC in vivo and in vitro following oestradiol administration. Concomitant changes in intracellular calcium levels have also been observed under the influence of oestradiol. To rule out the effects of cytokines, parallel studies were performed using primary cultures of VEC. Oestradiol-induced calcium uptake was seen even in the presence of actinomycin D and cycloheximide which inhibit transcription and translation respectively. Calcium uptake was blocked by neomycin, an inhibitor of phosphoinositol lipid metabolism, and by the oestrogen receptor antagonist tamoxifen. Results suggest that oestradiol induces second messenger pathways in the VEC through specific receptors. Implications of these observations in cellular differentiation processes are discussed.


Subject(s)
Epithelial Cells/drug effects , Estradiol/pharmacology , Phosphatidylinositols/pharmacology , Signal Transduction/drug effects , Vagina/cytology , Animals , Calcium/metabolism , Calcium/pharmacokinetics , Calcium/pharmacology , Cells, Cultured , Epithelial Cells/cytology , Female , Inositol/metabolism , Phosphatidylinositols/metabolism , Phospholipids/metabolism , Rats , Rats, Wistar , Receptors, Estradiol/physiology , Second Messenger Systems/physiology , Signal Transduction/physiology , Steroids/pharmacology , Tritium , Vagina/drug effects
16.
J Endocrinol ; 109(2): 251-5, 1986 May.
Article in English | MEDLINE | ID: mdl-3711762

ABSTRACT

Functional abnormalities have been observed previously in the oestrogen-responsive tissues of ovariectomized ewes with permanent infertility resulting from prolonged exposure to oestrogenic pastures. In the present study, such ewes had higher rates of protein and glycoprotein synthesis in the cervix and uterus than control ewes, but the number of oestradiol-17 beta receptors in the nucleus was similar in each group. After treatment with oestradiol-17 beta, the increase in synthesis of protein and glycoprotein in the uterus and cervix was less in clover-affected ewes, but the amount of oestradiol-17 beta-receptor complex in the nucleus of uterine cells 6 h after ewes were injected with oestradiol-17 beta was similar in clover-affected and control ewes. The rate of replenishment of oestradiol-17 beta receptors in the cytoplasm at 24 and 48 h after oestradiol-17 beta injection was also similar in both groups. The abnormal function seen in the genital tract of clover-affected ewes could not, therefore, be shown to depend on changes in oestradiol-17 beta receptors.


Subject(s)
Estrogens/physiology , Infertility, Female/veterinary , Receptors, Estradiol/physiology , Receptors, Estrogen/physiology , Sheep Diseases/physiopathology , Animals , Cell Nucleus/metabolism , Cervix Uteri/metabolism , Cytoplasm/analysis , Cytosol/metabolism , Female , Glycoproteins/biosynthesis , Ovariectomy , Protein Biosynthesis , Receptors, Estradiol/analysis , Sheep , Uterus/metabolism
17.
Mol Cell Endocrinol ; 43(2-3): 135-40, 1985 Dec.
Article in English | MEDLINE | ID: mdl-3935499

ABSTRACT

The present study examined the effects of streptozotocin-induced diabetes on prolactin (Prl) secretion and its correlation with estrogen receptor levels in the anterior pituitary and hypothalamus. Prl was measured in adult ovariectomized rats and after estradiol treatment (10 micrograms estradiol benzoate (Eb) 48, 24 and 1 h before experiments) or acute TRH administration (4 micrograms/kg body weight). Substantial decreases in estradiol- and TRH-induced Prl release were observed in diabetic rats. Insulin therapy was able to restore this response. Measurement of nuclear estradiol receptors by exchange assay in the pituitary of Eb-treated rats revealed a significant reduction in receptor levels in the diabetic group and a restoration to normal values in insulin-treated diabetic rats. Similar results were obtained by measuring total pituitary receptor content (cytosolic plus nuclear receptors). No significant changes were observed in nuclear hypothalamic estradiol receptors. However, the number of total hypothalamic estradiol receptors was diminished in diabetic rats although the translocation was proportionally greater in these animals. These results indicate that the disrupted reproductive functions described in streptozotocin diabetic rats may be due, at least in part, to deficiencies in Prl secretion and pituitary estradiol action.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Pituitary Gland/metabolism , Pituitary Gland/ultrastructure , Prolactin/metabolism , Receptors, Estradiol/physiology , Receptors, Estrogen/physiology , Animals , Estradiol/therapeutic use , Female , Insulin/physiology , Ovariectomy , Rats , Rats, Inbred Strains , Thyrotropin-Releasing Hormone/physiology
18.
Mol Cell Endocrinol ; 124(1-2): 71-7, 1996 Nov 29.
Article in English | MEDLINE | ID: mdl-9027326

ABSTRACT

The mRNA levels of LIV-1 and pS2, two estrogen-responsive genes, are increased by the agents, cholera toxin (CT) plus 3-isobutyl-l-methylxanthine (IBMX), which cause an increase in cAMP in MCF-7 human breast cancer cells. The simultaneous addition of estradiol and CT/IBMX results in a synergistic induction of the two mRNAs. The changes in mRNA reflect changes in transcription of the two genes. Interestingly, the addition of CT/IBMX to estradiol not only causes a greater increase in transcription rate but the increase is longer-lasting that seen with the hormone alone. Stimulation of mRNA levels by CT/IBMX, but not by estradiol, was prevented by cycloheximide. Stimulation by both estradiol and by CT/IBMX was prevented by the antiestrogen, ICI 164387. Transcription of LIV-1 and pS2 genes is by both estradiol and cAMP, via separate mechanisms both requiring the estrogen receptor.


Subject(s)
Cyclic AMP/physiology , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Neoplasm Proteins/genetics , Proteins , 1-Methyl-3-isobutylxanthine/pharmacology , Breast Neoplasms , Carcinoma , Cholera Toxin/pharmacology , Cycloheximide/pharmacology , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphodiesterase Inhibitors/pharmacology , Polyunsaturated Alkamides , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Estradiol/physiology , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
19.
Am J Clin Pathol ; 83(4): 444-9, 1985 Apr.
Article in English | MEDLINE | ID: mdl-3984938

ABSTRACT

Specific estrogen and progesterone binding in human endometrium was studied histochemically using fluorochrome-labeled steroids (estradiol-17 beta-BSA-FITC and progesterone-BSA-TMRITC), endometrial samples from 36 women being investigated. The binding pattern was similar with both reagents. The relationship between the bindings to glands and to stroma, however, varied with the menstrual phase of the tissue. The specific fluorescence was more intense in the epithelial structures in the proliferative phase. In the secretory phase, the fluorescence from stromal cells was as intense as, or more intense than, that from the glands. The localization of the fluorophores in the glandular epithelial cells also varied by menstrual phase. In the proliferative phase, the fluorescence was most intense in the basal part or the whole cytoplasm of the glandular epithelial cells, while in the secretory phase the fluorescence was most intense in the apical and sometimes also in the basal part of the epithelial cells.


Subject(s)
Endometrium/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Menstrual Cycle , Receptors, Estradiol/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Adult , Binding Sites , Endometrium/physiology , Estrogens/blood , Female , Fluoresceins , Humans , Progesterone/blood , Receptors, Estradiol/physiology , Receptors, Progesterone/physiology , Rhodamines , Serum Albumin, Bovine
20.
Behav Neurosci ; 106(1): 162-71, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1554429

ABSTRACT

The effects of intracranial implants of estradiol in the ventromedial hypothalamus (VMH), the anterior hypothalamus (AH), or the medial amygdala (AMG) on aggression, sexual behavior, and serum estradiol were examined in female Syrian hamsters. Estradiol implants in the VMH, followed by systemic progesterone, stimulated sexual behavior and inhibited aggression. Estradiol implants in other intracranial sites activated sexual behavior but did not reliably inhibit aggression. Intracranially implanted and systemically treated animals had equivalent peripheral estradiol concentrations at sacrifice. These results suggest that: (a) the VMH is an important neural site for estradiol actions on sexual and aggressive behavior, (b) the caudal AH and AMG also may be sites of estradiol action on sexual behavior, and (c) these intracranial implants may only be effective given systemic estradiol exposure or the concurrent stimulation of multiple brain areas.


Subject(s)
Aggression/physiology , Amygdala/physiology , Estradiol/physiology , Hypothalamus, Anterior/physiology , Receptors, Estradiol/physiology , Sexual Behavior, Animal/physiology , Ventromedial Hypothalamic Nucleus/physiology , Agonistic Behavior/physiology , Animals , Brain Mapping , Cricetinae , Female , Progesterone/physiology , Social Environment
SELECTION OF CITATIONS
SEARCH DETAIL