Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Neurobiol ; 40(5): 711-723, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31784921

ABSTRACT

It is known that spontaneously hypertensive rats (SHR) present a marked encephalopathy, targeting vulnerable regions such as the hippocampus. Abnormalities of the hippocampus of SHR include decreased neurogenesis in the dentate gyrus (DG), partial loss of neurons in the hilus of the DG, micro and astrogliosis and inflammation. It is also known that 17ß-estradiol (E2) exert neuroprotective effects and prevent hippocampal abnormalities of SHR. The effects of E2 may involve a variety of mechanisms, including intracellular receptors of the ERα and ERß subtypes or membrane-located receptors, such as the G protein-coupled estradiol receptor (GPER). We have now investigated the protective role of GPER in SHR employing its synthetic agonist G1. To accomplish this objective, 5 month-old male SHR received 150 µg/day of G1 during 2 weeks. At the end of this period, we analyzed neuronal progenitors by staining for doublecortin (DCX), and counted the number of glial fibrillary acidic protein (GFAP)-labeled astrocytes and Iba1-stained microglial cells by computerized image analysis. We found that G1 activation of GPER increased DCX+ cells in the DG and reduced GFAP+ astrogliosis and Iba1+ microgliosis in the CA1 region of hippocampus. We also found that the high expression of proinflammatory makers IL1ß and cyclooxygenase 2 (COX2) of SHR was decreased after G1 treatment, which correlated with a change of microglia phenotype from the activated to a resting morphology. Additionally, G1 treatment increased the anti-inflammatory factor TGFß in SHR hippocampus. Altogether, our results suggest that activation of GPER plays a neuroprotective role on the encephalopathy of SHR, an outcome resembling E2 effects but avoiding secondary effects of the natural hormone.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hippocampus/abnormalities , Hippocampus/pathology , Hypertensive Encephalopathy/metabolism , Inflammation/metabolism , Neurogenesis , Receptors, G-Protein-Coupled/metabolism , Animals , Astrocytes/metabolism , Doublecortin Protein , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Glial Fibrillary Acidic Protein , Hypertensive Encephalopathy/drug therapy , Male , Microglia/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Rats , Rats, Inbred SHR , Receptors, Estradiol/agonists , Receptors, Estradiol/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
2.
Prostaglandins Other Lipid Mediat ; 121(Pt B): 170-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26319698

ABSTRACT

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.


Subject(s)
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
3.
Reprod Biol Endocrinol ; 12: 90, 2014 Sep 20.
Article in English | MEDLINE | ID: mdl-25239217

ABSTRACT

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.


Subject(s)
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
4.
J Vet Med Sci ; 76(12): 1623-5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25649945

ABSTRACT

STX is an agonist for a recently characterized membrane estrogen receptor whose structure has not been identified. We evaluated whether STX suppresses gonadotropin-releasing hormone (GnRH)-induced luteinizing hormone (LH) release from bovine anterior pituitary (AP) cells. We cultured AP cells (n=12) for 3 days in steroid-free conditions, followed by increasing concentrations (0.001, 0.01, 0.1, 1 and 10 nM) of 17ß-estradiol or STX for 5 min before GnRH stimulation until the end of the experiment. Estradiol (0.001 to 0.1 nM) significantly suppressed GnRH-stimulated LH secretion, whereas STX did not affect GnRH-stimulated LH secretion at any of the tested concentrations. In conclusion, STX, unlike estradiol, possesses no suppressive effect on GnRH-induced LH release from bovine AP cells.


Subject(s)
Acrylamide/pharmacology , Biphenyl Compounds/pharmacology , Cells, Cultured/metabolism , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Pituitary Gland, Anterior/cytology , Receptors, Estradiol/agonists , Animals , Cattle , Dose-Response Relationship, Drug , Estradiol/pharmacology , Pituitary Gland, Anterior/metabolism
5.
Microvasc Res ; 85: 118-27, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23063870

ABSTRACT

The steroid hormone estradiol is suggested to play a protective role in intestinal injury during systemic inflammation (sepsis). Our aim was to determine the effects of specific estradiol receptor (ER-α and ER-ß) agonists on the intestinal microcirculation during experimental sepsis. Male and sham ovariectomized female rats were subjected to sham colon ascendens stent peritonitis (CASP), and they were compared to male and ovariectomized female rats underwent CASP and either estradiol receptor α (ER-α) agonist propyl pyrazole triol (PPT), estradiol receptor ß (ER-ß) agonist diarylpropiolnitrile (DPN), or vehicle treatment. Intravital microscopy was performed, which is sufficiently sensitive to measure changes in the functional capillary density (FCD) as well as the major steps in leukocyte recruitment (rolling and adhesion). The leukocyte extravasations were also quantified by using histological paraffin sections of formalin fixed intestine. We found that either DPN (ER-ß) or PPT (ER-α) significantly reduced (P<0.05) sepsis-induced leukocyte-endothelial interaction (rolling, adherent leukocytes and neutrophil extravasations) and improved the intestinal muscular FCD. [PPT: Female; Leukocyte rolling (n/min): V(3) 3.7±0.7 vs 0.8±0.2, Leukocyte adhesion(n/mm(2)): V(3) 131.3±22.6 vs 57.2±13.5, Neutrophil extravasations (n/10000 µm(2)): 3.1±0.7 vs 6 ±1. Male; Leukocyte adhesion (n/mm(2)): V(1) 154.8±19.2 vs 81.3±11.2, V(3) 115.5±23.1 vs 37.8±12]. [DPN: Female; neutrophil extravasations (n/10000 µm(2)) 3.8±0.6 vs 6 ±1. Male; Leukocyte adhesion (n/mm(2)) V(1) 154.8±19.2 vs 70±10.5, V(3) 115.5±23.1 vs 52.8±9.6].Those results suggest that the observed effects of estradiol receptors on different phases of leukocytes recruitment with the improvement of the functional capillary density could partially explain the previous demonstrated salutary effects of estradiol on the intestinal microcirculation during sepsis. The observed activity of this class of compounds could open up a new avenue of research into the potential treatment of sepsis.


Subject(s)
Microcirculation/physiology , Receptors, Estradiol/metabolism , Sepsis/metabolism , Animals , Blood Pressure , Cell Adhesion , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Heart Rate , Leukocyte Rolling/physiology , Leukocytes/cytology , Male , Microscopy/methods , Microscopy, Fluorescence/methods , Neutrophils/metabolism , Peritonitis/pathology , Rats , Rats, Inbred Lew , Receptors, Estradiol/agonists , Stents
6.
Bioorg Med Chem Lett ; 19(4): 1250-3, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19167882

ABSTRACT

Estradiol and related estrogens have been widely used as supplements to relieve menopausal symptoms, but they lead to an increased risk of breast and endometrial cancer. Here we report the synthesis of a new family of compounds where we have removed the B-ring from the steroid ABCD structure, and functionalized the A-ring. These A-CD compounds show a preferential affinity for the estrogen receptor subtype ERbeta. Some show binding affinities which are greater than estradiol. The presence of electron-withdrawing substituents on the A-ring should reduce the tendency of these compounds to form carcinogenic metabolites, so they might lead to a safer approach to hormone replacement therapy.


Subject(s)
Estradiol , Receptors, Estradiol/agonists , Estradiol/agonists , Estradiol/analogs & derivatives , Estradiol/chemical synthesis , Estradiol/pharmacology , Female , Hormone Replacement Therapy/methods , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
7.
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
8.
J Toxicol Environ Health ; 52(3): 189-209, 1997 Oct 24.
Article in English | MEDLINE | ID: mdl-9316643

ABSTRACT

Several strains of laboratory rats have a high background incidence of mammary tumors and develop a persistent, anovulatory estrus condition at about 12 mo of age. The increased tumor incidence is believed to be associated with elevated estradiol (E2) and prolactin during the period of persistent estrus. A pharmacodynamic estrus cycle (PD-EC) model for the Sprague-Dawley rats has been developed in an attempt to analyze the physiological basis of early-onset persistent estrus and to examine the potential sites of interactions in the hypothalamic-pituitary-ovarian axis for endocrine-modulating xenobiotics that accelerate the onset of persistent estrus. This initial estrus cycle model focused solely on cyclical changes in E2 and luteinizing hormone (LH). An LH surge was scheduled when a hypothetical estrus cycle-related protein (EC-RP) under transcriptional control by the E2 receptor reached a critical concentration. In the model, aging-related cumulative hypothalamic E2 exposure impaired the LH surge by reducing the rate of production of the EC-RP. The progressively decreasing intercycle resynthesis rate leads first to longer, variable-length cycles and finally to persistent estrus at about 12 mo of age. This model construct is consistent with early-onset persistent estrus related to neonatal E2 exposures, with acyclicity associated with high-dose E2 exposure in the adult, and with persistent estrus conditions associated with exposures to xenobiotic endocrine modulators that are either weak E2 antagonists or weak E2 agonists. With further development these pharmacodynamic estrus cycle models should be useful in aiding risk assessments for compounds causing mammary-tissue tumors associated with persistent estrus states.


Subject(s)
Estradiol/blood , Estrus/drug effects , Luteinizing Hormone/blood , Prolactin/blood , Receptors, Estradiol/agonists , Receptors, Estradiol/antagonists & inhibitors , Aging/drug effects , Animals , Chlordecone/toxicity , Dose-Response Relationship, Drug , Estrus/blood , Estrus/physiology , Female , Hypothalamus/drug effects , Hypothalamus/physiology , Insecticides/toxicity , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/physiopathology , Ovary/drug effects , Ovary/physiology , Pituitary Gland/drug effects , Pituitary Gland/physiology , Rats , Rats, Sprague-Dawley/genetics , Receptors, Estradiol/biosynthesis , Reproduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...