Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Neuroscience ; 426: 59-68, 2020 02 01.
Article En | MEDLINE | ID: mdl-31805254

Estradiol not only participates in the regulation of energy metabolism in adulthood, but also during the first stages of life as it modulates the alterations induced by under- and over-nutrition. The objectives of the present study were to determine: 1) If estradiol is involved in the normal programming of energy metabolism in rats; 2) If there is a specific window of time for this programming and 3) If males and females are differentially vulnerable to the action of this hormone. Estrogen receptors (ER) α, ERß and GPER were blocked by their specific antagonists MPP, PHTPP and G15, respectively, from postnatal day (P) 1 (the day of birth) to P5 or from P5 to P13. Physiological parameters such as body weight, fat depots and caloric intake were then analysed at P90. Hypothalamic AgRP, POMC, MC4R, ERα, ERß and GPER mRNA levels and plasma levels of estradiol, were also studied. We found that blocking ER receptors from P5 to P13 significantly decreases long-term body weight in males and hypothalamic POMC mRNA levels in females. The blocking of ERs from P1 to P5 only affected plasma estradiol levels in females. The present results indicate programming actions of estradiol from P5 to P13 on body weight in male and POMC expression in female rats and emphasize the importance of including both sexes in metabolic studies. It is necessary to unravel the mechanisms that underlie the actions of estradiol on food intake, both during development and in adulthood, and to determine how this programming differentially takes place in males and females.


Energy Metabolism/drug effects , Estrogen Receptor beta/antagonists & inhibitors , Receptors, Estradiol/antagonists & inhibitors , Animals , Body Weight/drug effects , Body Weight/physiology , Energy Metabolism/physiology , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Hypothalamus/metabolism , Male , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
2.
eNeuro ; 6(1)2019.
Article En | MEDLINE | ID: mdl-30766916

Estradiol potentiates behavioral sensitization to cocaine as well as self-administration of cocaine and other drugs of abuse in female rodents. Furthermore, stimulated dopamine (DA) in the dorsolateral striatum (DLS) is rapidly enhanced by estradiol, and it is hypothesized that this enhanced DA release mediates the more rapid escalation of drug taking seen in females, compared with males. The mechanisms mediating the effect of estradiol to enhance stimulated DA release were investigated in this study. Using in vivo microdialysis and high performance liquid chromatography coupled with electrochemical detection, we first examined the effect of estradiol on amphetamine-induced DA increase in the DLS of ovariectomized rats. We then tested whether the potentiation of this DA increase could be blocked by the estradiol receptor antagonist, ICI 182,780 (ICI), or an antagonist to the metabotropic glutamate receptor subtype 5 (mGlu5), 2-methyl-6-(phenylethynyl)pyridine (MPEP). There is evidence that estradiol receptors collaborate with mGlu5 within caveoli in DLS and mGlu5 is hypothesized to mediate many of the effects of estradiol in the addiction processes in females. Our data show that estradiol enhances the DA response to amphetamine. Either ICI or MPEP prevented the effect of estradiol to enhance DA release. Importantly, our results also showed that neither ICI or MPEP alone is able to influence the DA response to amphetamine when estradiol is not administrated, suggesting that ICI and MPEP act via estradiol receptors. Together, our findings demonstrate that estradiol potentiates amphetamine-stimulated DA release in the DLS and this effect requires both estradiol receptors and mGlu5.


Amphetamine/pharmacology , Central Nervous System Agents/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Estradiol/metabolism , Amphetamine-Related Disorders/metabolism , Animals , Estradiol/pharmacology , Female , Fulvestrant/pharmacology , Pyridines/pharmacology , Random Allocation , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/metabolism
3.
Prostaglandins Other Lipid Mediat ; 121(Pt B): 170-5, 2015 Sep.
Article En | MEDLINE | ID: mdl-26319698

Gamete and embryo transport is an important function of the oviduct. This transport involves both smooth muscle contraction and epithelial cell secretions, the former of which is mediated by prostaglandins (PGs) and their receptors. Our aim was to study the regulation of prostaglandin E2 and prostaglandin F2α receptors (EP2, EP4, and FP receptor) by estradiol in bovine oviduct smooth muscle. EP2, EP4, and FP receptor mRNA and protein expression was investigated using real-time RT-PCR and Western blot analyses, respectively. To evaluate the contraction or relaxation of cultured bovine oviductal smooth muscle tissue, peristalsis was used to assess contractile activity. EP2, EP4, and FP receptor mRNA and protein expression was increased in oviductal smooth muscle tissue after treatment with different concentrations of estradiol for various durations. The expression of all receptors peaked at an estradiol concentration of 10(-11)mol/L after 8h of treatment, whereas no increase in expression was observed after fulvestrant (a selective antagonist of E2 receptor) treatment, indicating that E2 interacts with specific E2 nuclear receptors to regulate EP2, EP4, and FP receptor expression. Although PGF2α and PGE2 induced both contraction and relaxation, no significant differences were found in contractility between the estradiol-treated and control groups, with both groups of cultured smooth muscle strips showing similar vitality. In conclusion, estradiol increases EP2, EP4, and FP receptor mRNA and protein expression in bovine oviductal smooth muscle when added for different periods of time and at different concentrations. Additionally, E2 is transported intracellularly and interacts with specific E2 nuclear receptors to regulate their expression.


Estradiol/metabolism , Muscle, Smooth/metabolism , Oviducts/metabolism , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin/agonists , Up-Regulation , Abattoirs , Absorption, Physiological/drug effects , Animals , Biological Transport/drug effects , Cattle , China , Crosses, Genetic , Estradiol/analogs & derivatives , Estradiol/chemistry , Estradiol/pharmacology , Estrogen Receptor Antagonists/pharmacology , Female , Fulvestrant , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Oviducts/drug effects , RNA, Messenger/metabolism , Receptors, Estradiol/agonists , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Tissue Culture Techniques , Up-Regulation/drug effects
4.
Reprod Biol Endocrinol ; 12: 90, 2014 Sep 20.
Article En | MEDLINE | ID: mdl-25239217

BACKGROUND: Aromatase converts testosterone into 17beta-estradiol in granulosa cells, and the converted 17beta-estradiol contributes to follicular maturation. Additionally, excessive testosterone inhibits aromatase activity, which can lead to concerns regarding polycystic ovary syndrome (PCOS). Generally, 1,25-dihydroxyvitamin D3 (1,25D3) supplements help to improve the symptoms of PCOS patients who exhibit low blood levels of 1,25D3. Therefore, this study investigated the interaction effects of 1,25D3 and testosterone on estrogenesis and intercellular connections in rat granulosa cells. METHODS: Primary cultures of granulosa cells were treated with testosterone or testosterone plus 1,25D3, or pre-treated with a calcium channel blocker or calcium chelator. Cell lysates were subjected to western blot analysis to determine protein and phosphorylation levels, and 17beta-estradiol secretion was examined using a radioimmunoassay technique. Cell viability was evaluated by MTT reduction assay. Connexin 43 (Cx43) mRNA and protein expression levels were assessed by qRT-PCR, western blot, and immunocytochemistry. RESULTS: Testosterone treatment (0.1 and 1 microg/mL) increased aromatase expression and 17beta-estradiol secretion, and the addition of 1,25D3 attenuated testosterone (1 microg/mL)-induced aromatase expression but improved testosterone-induced 17beta-estradiol secretion. Furthermore, testosterone-induced aromatase phosphotyrosine levels increased at 10 min, 30 min and 1 h, whereas 1,25D3 increased the longevity of the testosterone effect to 6 h and 24 h. Within 18-24 h of treatment, 1,25D3 markedly enhanced testosterone-induced 17beta-estradiol secretion. Additionally, pre-treatment with a calcium channel blocker nifedipine or an intracellular calcium chelator BAPTA-AM reduced 1,25D3 and testosterone-induced 17beta-estradiol secretion. Groups that underwent testosterone treatment exhibited significantly increased estradiol receptor beta expression levels, which were not affected by 1,25D3. Neither testosterone nor 1,25D3 altered 1,25D3 receptor expression. Finally, at high doses of testosterone, Cx43 protein expression was decreased in granulosa cells, and this effect was reversed by co-treatment with 1,25D3. CONCLUSIONS: These data suggest that 1,25D3 potentially increases testosterone-induced 17beta-estradiol secretion by regulating aromatase phosphotyrosine levels, and calcium increase is involved in both 1,25D3 and testosterone-induced 17beta-estradiol secretion. 1,25D3 reverses the inhibitory effect of testosterone on Cx43 expression in granulosa cells.


Calcitriol/metabolism , Connexin 43/metabolism , Estradiol/metabolism , Gene Expression Regulation, Developmental , Granulosa Cells/metabolism , Testosterone/metabolism , Up-Regulation , Animals , Aromatase/chemistry , Aromatase/metabolism , Calcium Channel Blockers/pharmacology , Calcium Chelating Agents/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Connexin 43/agonists , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Down-Regulation/drug effects , Estradiol/agonists , Estradiol/chemistry , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Granulosa Cells/cytology , Granulosa Cells/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats, Sprague-Dawley , Receptors, Estradiol/agonists , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/metabolism , Testosterone/agonists , Testosterone/antagonists & inhibitors , Up-Regulation/drug effects
5.
Am J Respir Cell Mol Biol ; 49(1): 135-42, 2013 Jul.
Article En | MEDLINE | ID: mdl-23526212

Lymphangioleiomyomatosis (LAM) is a destructive lung disease primarily affecting women. Genetic studies indicate that LAM cells carry inactivating tuberous sclerosis complex (TSC)-2 mutations, and metastasize to the lung. We previously discovered that estradiol increases the metastasis of TSC2-deficient cells in mice carrying xenograft tumors. Here, we investigate the molecular basis underlying the estradiol-induced lung metastasis of TSC2-deficient cells, and test the efficacy of Faslodex (an estrogen receptor antagonist) in a preclinical model of LAM. We used a xenograft tumor model in which estradiol induces the lung metastasis of TSC2-deficient cells. We analyzed the impact of Faslodex on tumor size, the extracellular matrix organization, the expression of matrix metalloproteinase (MMP)-2, and lung metastasis. We also examined the effects of estradiol and Faslodex on MMP2 expression and activity in tuberin-deficient cells in vitro. Estradiol resulted in a marked reduction of Type IV collagen deposition in xenograft tumors, associated with 2-fold greater MMP2 concentrations compared with placebo-treated mice. Faslodex normalized the Type IV collagen changes in xenograft tumors, enhanced the survival of the mice, and completely blocked lung metastases. In vitro, estradiol enhanced MMP2 transcripts, protein accumulation, and activity. These estradiol-induced changes in MMP2 were blocked by Faslodex. In TSC2-deficient cells, estradiol increased MMP2 concentrations in vitro and in vivo, and induced extracellular matrix remodeling. Faslodex inhibits the estradiol-induced lung metastasis of TSC2-deficient cells. Targeting estrogen receptors with Faslodex may be of efficacy in the treatment of LAM.


Estradiol/analogs & derivatives , Estradiol/adverse effects , Extracellular Matrix/drug effects , Lung Neoplasms/secondary , Lymphangioleiomyomatosis/pathology , Airway Remodeling , Animals , Antineoplastic Agents/pharmacology , Collagen Type IV/metabolism , Drug Evaluation, Preclinical , Estradiol/pharmacology , Extracellular Matrix/metabolism , Female , Fulvestrant , Lung/drug effects , Lung/metabolism , Lung/pathology , Lymphangioleiomyomatosis/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, SCID , Rats , Receptors, Estradiol/antagonists & inhibitors , Survival Analysis , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
6.
Endocrinology ; 152(3): 773-81, 2011 Mar.
Article En | MEDLINE | ID: mdl-21248144

Accumulating evidence suggests an enhancing effect of estrogens on prostate cancer (PCa) progression. Matrix metalloproteinase 2 (MMP2), which plays an important role in prostate cancer invasion, is mainly expressed in prostatic stromal cells (PrSC). Here we show that estradiol (E(2)) treatment up-regulates MMP2 production in PrSC, which promotes PCa cell invasion in a paracrine manner. Conditioned medium (CM) was collected from E(2)-treated prostatic stromal cell line WPMY-1 and primary PrSC. The CM of E(2)-treated WPMY-1 and PrSC promoted invasion of PCa cells, as measured by Matrigel transwell assays. Treatment with E(2) and 1,3,5-Tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole, an estrogen receptor-alpha (ERα) specific agonist, significantly up-regulated MMP2 expression in WPMY-1 and PrSC cells at both mRNA and protein levels. The CM treated with an anti-MMP2 antibody lost the stimulatory effect on invasion of PCa cells. The ER inhibitor ICI 182,780, as well as a TGFß1 neutralizing antibody and ERα-specific small interfering RNA effectively suppressed E(2)-induced MMP2 expression in WPMY-1 cells. Mechanistic studies showed that E(2) up-regulated MMP2 in an indirect manner: E(2) induced TGFß1 expression via ERα; TGFß1 stimulated MMP2 expression in PrSC; the invasion of PCa cells were stimulated by elevated MMP2 expression induced by E(2) in a paracrine manner. Our data show that E(2) induces MMP2 expression in WPMY-1 and PrSC cells, which was mediated by TGFß1. The effect of E(2) on invasion of PCa cells is mediated by up-regulation of MMP2 in a paracrine mechanism.


Estradiol/pharmacology , Matrix Metalloproteinase 2/metabolism , Paracrine Communication/physiology , Prostate/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Cell Line, Tumor , Culture Media, Conditioned , Estradiol/analogs & derivatives , Fulvestrant , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Matrix Metalloproteinase 2/genetics , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Receptors, Estradiol/antagonists & inhibitors , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
7.
Int Immunopharmacol ; 11(6): 675-82, 2011 Jun.
Article En | MEDLINE | ID: mdl-21262190

Cigarette smoking is highly associated with increased cardiovascular disease complications. The female population, however, manifests reduced cardiovascular morbidity. We define nicotine's effect upon human umbilical vein endothelial cells (HUVECs), determine whether estradiol might ameliorate endothelial dysfunction via its membrane estrogen receptor (mER), and attempt to elucidate the underlying mechanisms. Endothelial cells were pretreated with estradiol-BSA and measured resultant ion flux across the cells via the patch clamp technique to assess mER is functionality. Estradiol-BSA administration was associated with 30% decreased nicotine-induced apoptosis and also attenuated nicotine-activated phosphorylation of p38 and ERK. Pretreatment of estradiol-BSA triggered a low calcium influx, suggesting ahead low influx calcium played a critical role in the underlying protective mechanisms of estradiol. Furthermore, this estradiol-BSA protection against apoptosis remained effective in the presence of tamoxifen, an intracellular estrogen receptor (iER) inhibitor. Additionally, tamoxifen did not abolish estradiol-BSA's inhibitory effect upon p38 and ERK's activation, giving evidence to the obligatory role of p38 and ERK signaling in the estradiol-BSA's anti-apoptotic action via mER. Our study provides evidence that nicotine enhances endothelial cell apoptosis, but estrogen exerts anti-apoptotic effect through its functional membrane estrogen receptor. Clinically, the nicotine in cigarettes might contribute to endothelial dysfunction, whereas ambient estradiol may provide cellular protection against nicotine-induced injury through its functional membrane receptor via MAPK pathway downregulation.


Cardiovascular Diseases/metabolism , Endothelium, Vascular/drug effects , Estradiol/pharmacology , Receptors, Estradiol/metabolism , Sex Factors , Apoptosis/drug effects , Calcium Signaling/drug effects , Cardiovascular Diseases/epidemiology , Cell Line , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Male , Nicotine/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Receptors, Estradiol/antagonists & inhibitors , Risk , Smoking/adverse effects , Tamoxifen/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Oncogene ; 26(46): 6619-29, 2007 Oct 11.
Article En | MEDLINE | ID: mdl-17486077

In human mammary and prostate cancer cells, steroid hormones or epidermal growth factor (EGF) trigger association of the androgen receptor (AR)-estradiol receptor (ER) (alpha or beta) complex with Src. This interaction activates Src and affects the G1 to S cell cycle progression. In this report, we identify the sequence responsible for the AR/Src interaction and describe a 10 amino-acid peptide that inhibits this interaction. Treatment of the human prostate or mammary cancer cells (LNCaP or MCF-7, respectively) with nanomolar concentrations of this peptide inhibits the androgen- or estradiol-induced association between the AR or the ER and Src the Src/Erk pathway activation, cyclin D1 expression and DNA synthesis, without interfering in the receptor-dependent transcriptional activity. Similarly, the peptide prevents the S phase entry of LNCaP and MCF-7 cells treated with EGF as well as mouse embryo fibroblasts stimulated with androgen or EGF. Interestingly, the peptide does not inhibit the S phase entry and cytoskeletal changes induced by EGF or serum treatment of AR-negative prostate cancer cell lines. The peptide is the first example of a specific inhibitor of steroid receptor-dependent signal transducing activity. The importance of these results is highlighted by the finding that the peptide strongly inhibits the growth of LNCaP xenografts established in nude mice.


Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Androgen/metabolism , src Homology Domains/physiology , Amino Acid Sequence , Androgen Receptor Antagonists , Animals , Breast Neoplasms/metabolism , Humans , Male , Mice , Peptides , Prostatic Neoplasms/metabolism , Protein Binding , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/metabolism , Signal Transduction , Tumor Cells, Cultured
9.
Carcinogenesis ; 28(7): 1606-12, 2007 Jul.
Article En | MEDLINE | ID: mdl-17272310

Animal studies demonstrated that females are more susceptible than males to benzo[a]pyrene (BaP)-induced toxicities, including lung carcinogenesis. Elevation of cyclooxygenase-2 (COX-2) expression has been shown to increase the risk of cancer development. BaP induces COX-2 expression, and an interaction between BaP and estrogen in relation to COX-2 expression is suspected. In the present study, 10 muM BaP alone only slightly increased COX-2 mRNA expression and 10 nM 17-beta estradiol (E(2)) alone slightly increased prostaglandin E2 (PGE2) secretion in human bronchial epithelial cells. However, co-treatment with BaP and E(2) potentiated COX-2 mRNA expression and significantly elevated PGE2 secretion. Utilizing specific inhibitors and reporter assays, we further investigated the potentiation mechanisms of E(2) on BaP-induced COX-2 expression. First, E(2) activated estrogen receptor to increase PGE2 secretion, which directly increased COX-2 expression. Second, E(2) potentiated BaP-induced nuclear factor-kappaB (NF-kappaB) activation, which regulates COX-2 expression. Third, although the aryl hydrocarbon receptor (AhR) did not play a role in BaP-induced COX-2 expression, the potentiation effect of E(2) itself was AhR dependent. We further demonstrated that BaP induced the production of genotoxic E(2) metabolites (2- and 4-hydroxyestradiols) via AhR-up-regulated cytochromes P450 1A1 and 1B1. These metabolites could directly activate NF-kappaB to further promote COX-2 mRNA expression in human lung epithelial cells. These findings were further supported by increased PGE2 secretion in rat lung slice cultures. Our findings that the BaP-E(2) interaction enhanced COX-2 expression and hydroxyestradiol accumulation in the media of cultivated lung cells and tissues provide the needed scientific basis for higher risk of BaP-associated lung cancer in females.


Air Pollutants/toxicity , Benzo(a)pyrene/toxicity , Carcinogens, Environmental/toxicity , Cyclooxygenase 2/metabolism , Estradiol/analogs & derivatives , Estradiol/toxicity , Membrane Proteins/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Bronchi/cytology , Cells, Cultured , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1 , Dinoprostone/metabolism , Drug Synergism , Enzyme Activation , Epithelial Cells/metabolism , Estradiol/biosynthesis , Estradiol/pharmacology , Estrogens, Catechol , Fulvestrant , Humans , In Vitro Techniques , Lung/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Estradiol/antagonists & inhibitors , Up-Regulation
10.
Prostaglandins Other Lipid Mediat ; 80(3-4): 155-64, 2006 Sep.
Article En | MEDLINE | ID: mdl-16939880

We investigated the regulation of cyclooxygenase-2 (COX-2) by 17-beta-estradiol (E2) in the rat oviduct. We observed that COX-2 is expressed mainly in proestrous and estrous stages, periods under estrogenic influence. While exogenous administration of E2 (1 microg/rat) significantly increased COX-2 protein levels, progesterone did not modify it. COX-2 was mainly localized on oviductal epithelial cells from estrogenized rat. Induction of COX-2 expression by E2 was partially reverted by tamoxifen (1 mg/rat), an E2 receptor antagonist. Estradiol treatment also increased prostaglandins (PGs) synthesis: 6-keto-PGF(1alpha) (40%), a stable metabolite of prostacyclin (PGI2), PGF(2alpha) (40%) and PGE2 (50%). Tamoxifen completely suppressed this enhancement. In order to discriminate which isoform of COX was implicated in the stimulatory effect of E2 on PGs synthesis, oviducts were preincubated with meloxicam (Melo: 10(-9)M) or NS-398 (10(-7)M), two selective COX-2 inhibitors. Both Melo and NS-398 abolished the increase of PGs synthesis stimulated by E2. All together, these data indicate that E2 could upregulate COX-2 expression and activity in the rat oviduct and that the stimulatory effect of E2 may be receptor-mediated.


Cyclooxygenase 2/metabolism , Estradiol/pharmacology , Oviducts/drug effects , Oviducts/enzymology , Animals , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/analysis , Cyclooxygenase 2 Inhibitors/pharmacology , Epithelium/drug effects , Epithelium/enzymology , Estrous Cycle/metabolism , Female , Immunohistochemistry , Meloxicam , Membrane Proteins/metabolism , Nitrobenzenes/pharmacology , Oviducts/metabolism , Pregnancy , Progesterone/pharmacology , Prostaglandins/biosynthesis , Rats , Rats, Wistar , Receptors, Estradiol/antagonists & inhibitors , Sulfonamides/pharmacology , Tamoxifen/pharmacology , Thiazines/pharmacology , Thiazoles/pharmacology , Up-Regulation/drug effects
11.
Mech Ageing Dev ; 125(10-11): 679-82, 2004.
Article En | MEDLINE | ID: mdl-15541763

Evidence is presented for a unified model for the reaction of antiestrogens with estrogen receptors that explains much of the unusual pharmacology of these clinically important agents. Agonist activity results from occupancy of the estradiol-binding (primary) site in the receptor and antagonism from the additional interaction with a secondary locus not recognized by hormone. In the case of type I antiestrogens, such as tamoxifen, this is weaker than primary site binding, so these substances are agonists at low concentrations and antagonists at higher levels. With type II antiestrogens, affinities for both sites are comparable, so one never has the agonist situation (only primary site occupied), and these agents are pure antagonists. Reproductive tissues of the mouse and guinea pig contain a small macromolecule that binds hydroxytamoxifen, but not estradiol, keeping the free antiestrogen concentration below that required for secondary binding. Thus, in these species, tamoxifen is a pure agonist.


Estrogen Antagonists/metabolism , Models, Biological , Receptors, Estradiol/metabolism , Animals , Estrogen Antagonists/pharmacokinetics , Female , Guinea Pigs , Mice , Receptors, Estradiol/antagonists & inhibitors
12.
Neurosci Lett ; 367(1): 60-5, 2004 Aug 26.
Article En | MEDLINE | ID: mdl-15308298

During inflammation, microglial cells go through phenotypic and functional changes that include the production and release of large amounts of oxygen and nitrogen radicals. As such, activated microglia are subject to heightened oxidative stress. The multicatalytic proteasome clears oxidized and damaged proteins from cells, and has been shown to be an important aspect of the microglial compensatory response to activation. The female sex steroid estrogen is both cytoprotective and anti-inflammatory, and has been shown to affect microglial signaling in particular. To determine if estrogen might affect the proteasome in microglial cells, we examined the effects of 17 beta-estradiol treatment on proteasome activity in N9 microglial cells. Specifically, we measured ATP-dependent and ATP-independent chymotrypsin-like, trypsin-like, and peptidyl glutamyl peptide hydrolase (PGPH)-like activities in response to both 17 beta-estradiol and interferon gamma. Data indicate that estrogen, but not interferon gamma, significantly increases ATP-dependent chymotrypsin-like and PGPH-like activity. Furthermore, this effect was blocked by the p44/42 MAPK inhibitor PD98059. Hence, these data demonstrate that through the MAPK pathway, estrogen can upregulate proteasome activity, suggesting a possible mechanism for estrogen's cytoprotective effects.


Estrogens/pharmacology , Microglia/drug effects , Proteasome Endopeptidase Complex/drug effects , Adenosine Triphosphate/pharmacology , Analysis of Variance , Animals , Cell Line , Chymotrypsin/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Interferon-gamma/metabolism , Leupeptins/pharmacology , Mice , Microglia/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Receptors, Estradiol/antagonists & inhibitors , Tetrazolium Salts , Thiazoles , Time Factors
13.
Biochem Cell Biol ; 82(3): 381-90, 2004 Jun.
Article En | MEDLINE | ID: mdl-15181472

17Beta-estradiol has potent Ca2+ ionophore capability and its signaling in macrophages is mediated through binding to surface and genomic receptors, resulting in transient nitric oxide (NO) elaboration. We decided to examine if the transient release of NO is due to Ca2+ influx pattern or the quenching effect of superoxide (*O2-) through peroxynitrite formation. Differential chelation of intracellular Ca2+ ([Ca2+]i) showed that NO generation was favored by [Ca2+]i concentration of 237 nM. Application of an estrogen receptor antagonist ICI 182 780 resulted in attenuation of estradiol mediated NO release. Studies directed at identifying the possible role of *O2-; in the attenuation of NO showed no supportive evidence. Inhibition of extracellular Ca2+ channel or extracellular and intracellular Ca2+ channels showed data consistent with a case for optimum Ca2+ influx signal favoring iNOS gene expression, accompanied by an elevation in iNOS protein. These data show that Ca2+ influx pattern determines macrophage NO elaboration.


Calcium Channels/metabolism , Calcium/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Macrophages/metabolism , Nitric Oxide Synthase/biosynthesis , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Cell Line , Estrogen Antagonists/pharmacology , Fulvestrant , Gene Expression Regulation, Enzymologic/drug effects , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II , Receptors, Estradiol/antagonists & inhibitors , Superoxides/metabolism
14.
J Neurosci ; 24(23): 5315-21, 2004 Jun 09.
Article En | MEDLINE | ID: mdl-15190103

Intracellular amyloidbeta peptide (iAbeta1-42) accumulates in the Alzheimer's disease brain before plaque and tangle formation (Gouras et al., 2000) and is extremely toxic to human neurons (Zhang et al., 2002). Here, we investigated whether androgen and estrogen could prevent iAbeta1-4) toxicity, because both these hormones have a wide range of neuroprotective actions. At physiological concentrations, 17-beta-estradiol, testosterone, and methyl testosterone reduce iAbeta1-42-induced cell death by 50% in neurons treated after the injection and by 80-90% in neurons treated 1 hr before the injection. The neuroprotective action of the hormones is mediated by receptors, because the estrogen receptor (ER) antagonist tamoxifen and the androgen receptor (AR) antagonist flutamide completely block the estrogen- and androgen-mediated neuroprotection, respectively. Transcriptional activity is required for the neuroprotective action, because dominant negative forms of the receptors that block the transcriptional activity of the ER and AR prevent estrogen- and androgen-mediated neuroprotection. Proteomics followed by Western blot analyses identified increased levels of heat shock protein 70 (Hsp70) in testosterone- and estrogen-treated human neurons. Comicroinjection of Hsp70 with the iAbeta1-42 blocks the toxicity of iAbeta1-42. We conclude that estrogen and androgens protect human neurons against iAbeta1-42 toxicity by increasing the levels of Hsp70 in the neurons.


Amyloid beta-Peptides/adverse effects , Cerebral Cortex/physiology , Estradiol/physiology , HSP70 Heat-Shock Proteins/metabolism , Neurons/physiology , Testosterone/physiology , Amyloid beta-Peptides/metabolism , Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Cell Culture Techniques , Cell Death/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , DNA, Complementary , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Humans , In Situ Nick-End Labeling , Neurons/metabolism , Neuroprotective Agents/pharmacology , Receptors, Estradiol/antagonists & inhibitors , Testosterone/metabolism , Testosterone/pharmacology , Transcription, Genetic
15.
Xenobiotica ; 34(3): 257-72, 2004 Mar.
Article En | MEDLINE | ID: mdl-15204698

1: The metabolism and action of chlorpropham (isopropyl N-(3-chlorophenyl)carbamate; CIPC, a post-harvest agent) were studied in freshly isolated rat hepatocytes, and the oestrogen-like activity of CIPC and its metabolites was assessed by in vitro assays. The exposure of hepatocyte suspensions to CIPC caused concentration- (0.25-1.0 mM) and time- (0-3 h) dependent cell death, which was assessed by Trypan blue exclusion, accompanied by losses of cellular adenosine triphosphate and adenine nucleotide pools, and formation of cell bleb. 2: CIPC at a weakly toxic level (0.25 or 0.5 mM) was metabolized to isopropyl N-(3-chloro-4-hydroxyphenyl)carbamate (4OH-CIPC) and subsequently to its glucuronide and sulfate conjugates (major metabolites) or alternatively to the minor metabolites 3-chloroaniline (3CA) and 3-chloroacetanilide. CIPC (0.25 mM) added to hepatocyte suspensions was distributed equally between hepatocytes and the extracellular medium during the incubation. The glucuronide rather than the sulfate conjugate of 4OH-CIPC predominantly increased in the medium with time, while the amount of unconjugated free 4OH-CIPC in the extracellular medium increased by approximately threefold compared with the amount in the cell fraction after 0.5 h and then decreased rapidly accompanied by increases in the conjugates. This indicates that unconjugated free 4OH-CIPC produced in hepatocytes was temporarily excreted in the extracellular medium and subsequently converted to the conjugates via re-influx into hepatocytes. 3: Diethylstilbestrol (DES), bisphenol A (BPA) and 4-hydroxybenzoic acid butyl ester (butylparaben), which are known xenoestrogenic compounds, competitively displaced 17beta-oestradiol bound to the oestrogen receptor-alpha (ERalpha) in a concentration-dependent manner; IC50 values of DES, BPA, butylparaben and its derivative 3-chloro-4-hydroxybenzoic acid butyl ester (3-chloro-butylparaben) were approximately 10(-8), 10(-5), 5 x 10(-5) and 5 x 10(-4) M, respectively. In contrast, neither CIPC nor 4OH-CIPC impaired the binding of 17beta-oestradiol to ERalpha at concentrations ranging from 10(-9) to 10(-4) M, whereas at concentrations of >5 x 10(-4) M, the binding affinity of 4OH-CIPC was greater than that of CIPC. In a proliferation assay of MCF-7 cells, CIPC, 4OH-CIPC and 3CA did not increase cell numbers at concentrations ranging from 10(-9) to 10(-5) M, but these compounds at a concentration of 10(-4) M induced a considerable decrease in cell numbers relative to the control. The results suggest that even if CIPC is metabolized to 4OH-CIPC by hepatocytes, the chlorine adjacent to the 4-hydroxy group added to the intermediate as well as 3-chloro-butylparaben obstructs the appearance of oestrogen-like effects via an interaction between the intermediate and the ER.


Chlorpropham/pharmacology , Estradiol/metabolism , Estrogens, Non-Steroidal/metabolism , Hepatocytes/metabolism , Herbicides/pharmacology , Receptors, Estradiol/metabolism , Animals , Biotransformation , Cell Death/drug effects , Cells, Cultured , Chlorpropham/metabolism , Chlorpropham/toxicity , Dose-Response Relationship, Drug , Hepatocytes/pathology , Herbicides/metabolism , Herbicides/toxicity , Male , Protein Binding , Rats , Rats, Inbred F344 , Receptors, Estradiol/antagonists & inhibitors
16.
Life Sci ; 71(1): 15-29, 2002 May 24.
Article En | MEDLINE | ID: mdl-12020745

This study investigated the effects of testosterone and 17-beta-estradiol on tumor necrosis factor-alpha (TNF-alpha)-induced endothelial expression of E-selectin and vascular cell adhesion molecule-1 (VCAM-1) and the potential roles of hormone receptors involved in these actions. Human umbilical vein endothelial cells (HUVEC) were stimulated with TNF-alpha in the presence or absence of testosterone or 17-beta-estradiol, and the expression of E-selectin and VCAM-1 was investigated. As shown by Western blot analysis, co-administration with testosterone or 17-beta-estradiol increased the expression of E-selectin and VCAM-1 induced by TNF-alpha at 6 h and 3 h, respectively. Similarly, RT-PCR analysis revealed a significant increase in the amount of mRNA for E-selectin and VCAM-1 after co-administration with testosterone or 17-beta-estradiol in TNF-alpha-stimulated HUVEC. The presence of mRNA and proteins for androgen receptor and estrogen receptor alpha in HUVEC was verified by RT-PCR and Western blot. Flow cytometric analysis showed that preincubation with androgen receptor antagonist cyproterone and estrogen receptor antagonist tamoxifen completely abrogated the upregulating effects of testosterone and 17-beta-estradiol on TNF-alpha-induced E-selectin and VCAM-1 expression, respectively. Expression of TNF receptors in TNF-alpha-stimulated HUVEC was not influenced by testosterone and 17-beta-estradiol. The data indicate that both testosterone and 17-beta-estradiol increase TNF-alpha-induced E-selectin and VCAM-1 expression in endothelial cells via a receptor-mediated system, and expression of TNF receptors are not changed in these actions. The implications of these results for the facilitory effects of both sex hormones on immune reactions are discussed.


E-Selectin/biosynthesis , Endothelium, Vascular/metabolism , Estradiol/pharmacology , Testosterone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Androgen Receptor Antagonists , Antigens, CD/biosynthesis , Antigens, CD/genetics , Blotting, Western , Cell Line , Depression, Chemical , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flow Cytometry , Humans , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/biosynthesis , Receptors, Androgen/drug effects , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/drug effects , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
17.
Autoimmunity ; 33(4): 237-43, 2001.
Article En | MEDLINE | ID: mdl-11683398

The effects of oestradiol and the oestrogen receptor antagonist ICI 182,780 were investigated on the DTH index, serum IgG and IgM levels and spleen weight in female BALB/c and MRLL/MP-lpr/lpr mice. At six weeks, the mice were ovariectomised, and one week later, over a four-week period, given biweekly s.c. doses of (i) 5 microl of olive oil, or (ii) 5 microl of oil containing 3.2 microg of 17beta-oestradiol (E2), or (iii) 5 microl of oil containing (3.2 microl of E2 + ICI 182,780, at a dose of 30 mg/kg), or (iv) the same dose of ICI 182,780 alone, E2 significantly raised the DTH index in BALB/c mice; this effect was prevented if ICI 182,780 was included in the injection. The DTH index in MRL mice was unaffected by any of the treatments. All steroid treatments raised serum IgG levels in BALB/c mice, but those in sera of MRL were unaffected and were significantly higher than those measured in BALB/c mice. ICI 182,780 depressed IgM in BALB/c mice, while all steroid treatments increased IgM in MRL mice. ICI 182,780 depressed spleen weights in both strains. Oestrogen may influence B cell function through ICI 182,780-sensitive receptors, and ICI 182,780 may have agonist actions on the immune system. (200 words).


Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Hypersensitivity, Delayed/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Estradiol/antagonists & inhibitors , Animals , Female , Fulvestrant , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Ovariectomy , Species Specificity
18.
Biotechniques ; 29(6): 1226-31, 2000 Dec.
Article En | MEDLINE | ID: mdl-11126125

We have refined the regulated expression of UASGAL1, 10-driven genes in yeast by modifying a vector encoding the beta-estradiol inducible activator, GAL4.ER.VP16 (GEV). The expression of GEV was placed under the regulation of the low-level, constitutive MRP7 promoter, and beta-estradiol-regulated expression was monitored by the expression of an integrated UASGAL10-lacZ reporter and by immunoblot analysis of a UASGAL1-regulated gene product. Target gene expression regulated by low levels of GEV has several advantages over the standard galactose-inducible expression systems. (i) Most importantly, the target gene expression is undetectable in the absence of hormone; (ii) target gene expression is beta-estradiol dose-dependent, and variable levels of target gene expression from low to several hundred-fold induction can be achieved; and (iii) induction or depletion studies can be conducted independent of carbon source in gal4 delta strains. In addition, any UASGAL1,10 expression construct can be used without modification of the target gene or many gal4 delta host strains, and GEV vectors are compatible with other inducible yeast expression systems. This method may be useful to researchers investigating the functions of essential genes, dominant negative mutants, mitochondrial genes, and viral, plant, and mammalian genes in yeast assay systems.


Estradiol/pharmacology , Fungal Proteins/genetics , Gene Expression Regulation, Fungal/drug effects , Gene Targeting , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Carbon/physiology , DNA-Binding Proteins , Dose-Response Relationship, Drug , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/biosynthesis , Genetic Vectors/chemical synthesis , Genetic Vectors/metabolism , Humans , Raffinose/physiology , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/biosynthesis , Receptors, Estradiol/genetics , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/virology , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis
19.
Biol Reprod ; 63(1): 259-66, 2000 Jul.
Article En | MEDLINE | ID: mdl-10859267

This study was conducted to determine if the cadmium-mediated inhibition of vitellogenesis observed in fish collected from contaminated areas or undergoing experimental exposure to cadmium correlated with modification in the transcriptional activity of the estrogen receptor. A recombinant yeast system expressing rainbow trout (Oncorhynchus mykiss) estradiol receptor or human estradiol receptor was used to evaluate the direct effect of cadmium exposure on estradiol receptor transcriptional activity. In recombinant yeast, cadmium reduced the estradiol-stimulated transcription of an estrogen-responsive reporter gene. In vitro-binding assays indicated that cadmium did not affect ligand binding to the receptor. Yeast one- and two-hybrid assays showed that estradiol-induced conformational changes and receptor dimerization were not affected by cadmium; conversely, DNA binding of the estradiol receptor to its cognate element was dramatically reduced in gel retardation assay. This study provides mechanistic data supporting the idea that cadmium is an important endocrine disrupter through a direct effect on estradiol receptor transcriptional activity and may affect a number of estrogen signaling pathways.


Cadmium/pharmacology , Oncorhynchus mykiss/metabolism , Receptors, Estrogen/antagonists & inhibitors , Animals , DNA/metabolism , Dimerization , Estradiol/metabolism , Furylfuramide/metabolism , Humans , Metals, Heavy/pharmacology , Oncorhynchus mykiss/genetics , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription, Genetic , Vitellogenins/drug effects , Vitellogenins/metabolism , Yeasts/genetics
20.
Can J Physiol Pharmacol ; 76(4): 381-6, 1998 Apr.
Article En | MEDLINE | ID: mdl-9795746

In previous studies, we have shown that the baroreflex control of heart rate is significantly attenuated in females compared with age-matched males. This study investigated the role of estrogen in the modulation of baroreflex function in conscious unrestrained rats. Baroreflex-mediated decreases in heart rate in response to increments in blood pressure evoked by phenylephrine were evaluated in conscious freely moving male and female Sprague-Dawley rats as well as in ovariectomized rats. The effect of a 2-day 17 beta-estradiol (50 micrograms.kg-1.day-1, s.c.) or vehicle treatment on baroreflex sensitivity was investigated in ovariectomized rats. Intravenous bolus doses of phenylephrine (1-16 micrograms/kg) elicited dose-dependent pressor and bradycardic responses in all groups of rats. Regression analysis of the baroreflex curves relating increments in blood pressure to the associated heart rate responses revealed a significantly (p < 0.05) smaller baroreflex sensitivity in female compared with male rats (-1.22 +/- 0.07 and -1.85 +/- 0.15 beats.min-1.mmHg-1, respectively), suggesting an attenuated baroreflex function in females. In age-matched ovariectomized rats, baroreflex sensitivity showed further reduction (-0.93 +/- 0.02 beats.min-1.mmHg-1). Treatment of ovariectomized rats with 17 beta-estradiol significantly (p < 0.05) enhanced the baroreflex sensitivity (-1.41 +/- 0.16 beats.min-1.mmHg-1) to a level that was slightly higher than that of sham-operated female rats. Furthermore, baroreflex sensitivity of ovariectomized estradiol-treated rats was not significantly different from that of age-matched male rats. The vehicle, on the other hand, had no effect on baroreflex sensitivity of ovariectomized rats. These data support our earlier findings that sexual dimorphism exists in baroreflex control of heart rate. More importantly, the present study provides experimental evidence that suggests a facilitatory role for estrogen in the modulation of baroreflex function.


Baroreflex/drug effects , Estradiol/pharmacology , Heart Rate/drug effects , Receptors, Estradiol/physiology , Animals , Baroreflex/physiology , Female , Heart Rate/physiology , Male , Ovariectomy , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estradiol/antagonists & inhibitors , Sex Factors
...