Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 489
1.
J Reprod Dev ; 69(6): 337-346, 2023 Dec 08.
Article En | MEDLINE | ID: mdl-37940556

Insulin-like growth factor-1 (IGF-1) plays a crucial role in follicular growth and stimulates steroid hormone production in bovine follicles. Steroid hormones are synthesized through the actions of steroidogenic enzymes, specifically STAR, CYP11A1, HSD3B, and CYP19A1 in both theca cells (TCs) and granulosa cells (GCs), under the influence of gonadotropins. Particularly, estradiol 17ß (E2) assumes a central role in follicular development and selection by activating estrogen receptors ß (ESR2) in GCs. We assessed ESR2 mRNA expression in GCs of developing follicles and investigated the impact of IGF-1 on the mRNA expression of ESR2, CYP19A1, FSHR, and LHCGR, STAR, CYP11A1, and HSD17B in cultured GCs and TCs, respectively. Additionally, we assessed the influence of IGF-1 on androstenedione (A4), progesterone (P4), and testosterone (T) production in TCs. Small-sized follicles (< 6 mm) exhibited the highest levels of ESR2 mRNA expression, whereas medium-sized follicles (7-8 mm) displayed higher levels than large-sized follicles (≥ 9 mm) (P < 0.05). IGF-1 increased the mRNA expression of ESR2, CYP19A1, and FSHR in GCs of follicles of both sizes, except for FSHR mRNA in medium-sized follicles (P < 0.05). IGF-1 significantly elevated mRNA expression of LHCGR, STAR, CYP11A1, and CYP17B in TCs of small- and medium-sized follicles (P < 0.05). Moreover, IGF-1 augmented the production of A4 and P4 but had no impact on T production in TCs of small- and medium-sized follicles. Taken together, our findings indicate that IGF-1 upregulates steroidogenic enzymes and steroid hormone production, underscoring the crucial role of IGF-1 in follicle development and selection.


Gonadal Steroid Hormones , Insulin-Like Growth Factor I , Ovarian Follicle , Animals , Cattle , Female , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Estradiol/metabolism , Granulosa Cells/metabolism , Insulin-Like Growth Factor I/metabolism , Ovarian Follicle/metabolism , Progesterone/pharmacology , Receptors, Estradiol/metabolism , RNA, Messenger/metabolism , Gonadal Steroid Hormones/metabolism
2.
Diabetes ; 72(10): 1433-1445, 2023 10 01.
Article En | MEDLINE | ID: mdl-37478155

Altered endoplasmic reticulum (ER) Ca2+ signaling has been linked with ß-cell dysfunction and diabetes development. Store-operated Ca2+ entry replenishes ER Ca2+ through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor, stromal interaction molecule 1 (STIM1). For characterization of the in vivo impact of STIM1 loss, mice with ß-cell-specific STIM1 deletion (STIM1Δß mice) were generated and challenged with high-fat diet. Interestingly, ß-cell dysfunction was observed in female, but not male, mice. Female STIM1Δß mice displayed reductions in ß-cell mass, a concomitant increase in α-cell mass, and reduced expression of markers of ß-cell maturity, including MafA and UCN3. Consistent with these findings, STIM1 expression was inversely correlated with HbA1c levels in islets from female, but not male, human organ donors. Mechanistic assays demonstrated that the sexually dimorphic phenotype observed in STIM1Δß mice was due, in part, to loss of signaling through the noncanonical 17-ß estradiol receptor (GPER1), as GPER1 knockdown and inhibition led to a similar loss of expression of ß-cell maturity genes in INS-1 cells. Together, these data suggest that STIM1 orchestrates pancreatic ß-cell function and identity through GPER1-mediated estradiol signaling. ARTICLE HIGHLIGHTS: Store-operated Ca2+ entry replenishes endoplasmic reticulum (ER) Ca2+ through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor, stromal interaction molecule 1 (STIM1). ß-Cell-specific deletion of STIM1 results in a sexually dimorphic phenotype, with ß-cell dysfunction and loss of identity in female but not male mice. Expression of the noncanonical 17-ß estradiol receptor (GPER1) is decreased in islets of female STIM1Δß mice, and modulation of GPER1 levels leads to alterations in expression of ß-cell maturity genes in INS-1 cells.


Calcium Channels , Membrane Proteins , Animals , Mice , Female , Humans , Membrane Proteins/metabolism , Calcium Channels/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Calcium/metabolism , Receptors, Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Calcium Signaling , GTP-Binding Proteins/metabolism
3.
J Neuroendocrinol ; 35(1): e13228, 2023 01.
Article En | MEDLINE | ID: mdl-36690381

Hippocampal neuropathology is a recognized feature of the spontaneously hypertensive rat (SHR). The hippocampal alterations associate with cognitive impairment. We have shown that hippocampal abnormalities are reversed by 17ß-estradiol, a steroid binding to intracellular receptors (estrogen receptor α and ß subtypes) or the membrane-located G-protein coupled estradiol receptor. Genistein (GEN) is a neuroprotective phytoestrogen which binds to estrogen receptor ß and G-protein coupled estradiol receptor. Here, we investigated whether GEN neuroprotection extends to SHR. For this purpose, we treated 5-month-old SHR for 2 weeks with 10 mg kg-1 daily s.c injections of GEN. We analyzed the expression of doublecortin+ neuronal progenitors, glial fibrillary acidic protein+ astrocytes and ionized calcium-binding adapter molecule 1+ microglia in the CA1 region and dentate gyrus of the hippocampus using immunocytochemistry, whereas a quantitative real-time polymerase chain reaction was used to measure the expression of pro- and anti-inflammatory factors tumor necrosis factor α, cyclooxygenase-2 and transforming growth factor ß. We also evaluated hippocampal dependent memory using the novel object recognition test. The results showed a decreased number of doublecortin+ neural progenitors in the dentate gyrus of SHR that was reversed with GEN. The number of glial fibrillary acidic protein+ astrocytes in the dentate gyrus and CA1 was increased in SHR but significantly decreased by GEN treatment. Additionally, GEN shifted microglial morphology from the predominantly activated phenotype present in SHR, to the more surveillance phenotype found in normotensive rats. Furthermore, treatment with GEN decreased the mRNA of the pro-inflammatory factors tumor necrosis factor α and cyclooxygenase-2 and increased the mRNA of the anti-inflammatory factor transforming growth factor ß. Discrimination index in the novel object recognition test was decreased in SHR and treatment with GEN increased this parameter. Our results indicate important neuroprotective effects of GEN at the neurochemical and behavioral level in SHR. Our data open an interesting possibility for proposing this phytoestrogen as an alternative therapy in hypertensive encephalopathy.


Genistein , Phytoestrogens , Rats , Animals , Rats, Inbred SHR , Genistein/pharmacology , Phytoestrogens/pharmacology , Phytoestrogens/metabolism , Glial Fibrillary Acidic Protein/metabolism , Receptors, Estradiol/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cyclooxygenase 2/metabolism , Rats, Inbred WKY , Hippocampus/metabolism , Transforming Growth Factor beta/metabolism , Doublecortin Domain Proteins , RNA, Messenger/metabolism
4.
PLoS One ; 17(6): e0270609, 2022.
Article En | MEDLINE | ID: mdl-35763527

Covid-19 progression shows sex-dependent features. It is hypothesized that a better Covid-19 survival rate in females can be attributed to the presence of higher 17ß-estradiol (E2) levels in women than in men. Virus SARS-CoV-2 is enabled to enter the cell with the use of angiotensin converting enzyme 2 (ACE2). The expression of several renin-angiotensin system components has been shown to exert a rhythmic pattern, and a role of the circadian system in their regulation has been implicated. Therefore, the aim of the study is to elucidate possible interference between E2 signalling and the circadian system in the regulation of the expression of ACE2 mRNA and functionally related molecules. E2 was administered at a dosage of 40 µg/kg/day for 7 days to male Wistar rats, and sampling of the lungs and colon was performed during a 24-h cycle. The daily pattern of expression of molecules facilitating SARS-CoV-2 entry into the cell, clock genes and E2 receptors was analysed. As a consequence of E2 administration, a rhythm in ACE2 and TMPRSS2 mRNA expression was observed in the lungs but not in the colon. ADAM17 mRNA expression showed a pronounced rhythmic pattern in both tissues that was not influenced by E2 treatment. ESR1 mRNA expression exerted a rhythmic pattern, which was diminished by E2 treatment. The influence of E2 administration on ESR2 and GPER1 mRNA expression was greater in the lungs than in the colon as a significant rhythm in ESR2 and GPER1 mRNA expression appeared only in the lungs after E2 treatment. E2 administration also increased the amplitude of bmal1 expression in the lungs, which implicates altered functioning of peripheral oscillators in response to E2 treatment. The daily pattern of components of the SARS-CoV-2 entrance pathway and their responsiveness to E2 should be considered in the timing of pharmacological therapy for Covid-19.


ADAM17 Protein , Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , COVID-19 , Colon , Estradiol , Lung , Receptors, Estradiol , ADAM17 Protein/genetics , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/virology , Colon/drug effects , Colon/metabolism , Estradiol/pharmacology , Female , Lung/metabolism , Male , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Estradiol/genetics , Receptors, Estradiol/metabolism , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Transcription, Genetic/drug effects , Virus Internalization
5.
Elife ; 112022 04 13.
Article En | MEDLINE | ID: mdl-35416774

Skeletal muscle plays an integral role in coordinating physiological homeostasis, where signaling to other tissues via myokines allows for coordination of complex processes. Here, we aimed to leverage natural genetic correlation structure of gene expression both within and across tissues to understand how muscle interacts with metabolic tissues. Specifically, we performed a survey of genetic correlations focused on myokine gene regulation, muscle cell composition, cross-tissue signaling, and interactions with genetic sex in humans. While expression levels of a majority of myokines and cell proportions within skeletal muscle showed little relative differences between males and females, nearly all significant cross-tissue enrichments operated in a sex-specific or hormone-dependent fashion; in particular, with estradiol. These sex- and hormone-specific effects were consistent across key metabolic tissues: liver, pancreas, hypothalamus, intestine, heart, visceral, and subcutaneous adipose tissue. To characterize the role of estradiol receptor signaling on myokine expression, we generated male and female mice which lack estrogen receptor α specifically in skeletal muscle (MERKO) and integrated with human data. These analyses highlighted potential mechanisms of sex-dependent myokine signaling conserved between species, such as myostatin enriched for divergent substrate utilization pathways between sexes. Several other putative sex-dependent mechanisms of myokine signaling were uncovered, such as muscle-derived tumor necrosis factor alpha (TNFA) enriched for stronger inflammatory signaling in females compared to males and GPX3 as a male-specific link between glycolytic fiber abundance and hepatic inflammation. Collectively, we provide a population genetics framework for inferring muscle signaling to metabolic tissues in humans. We further highlight sex and estradiol receptor signaling as critical variables when assaying myokine functions and how changes in cell composition are predicted to impact other metabolic organs.


The muscles that are responsible for voluntary movements such as exercise are called skeletal muscles. These muscles secrete proteins called myokines, which play roles in a variety of processes by interacting with other tissues. Essentially, myokines allow skeletal muscles to communicate with organs such as the kidneys, the liver or the brain, which is essential for the body to keep its metabolic balance. Some of the process myokines are involved include inflammation, cancer, the changes brought about by exercise, and even cognition. Despite the clear relevance of myokines to so many physiological outcomes, the way these proteins are regulated and their effects are not well understood. Genetic sex ­ specified by sex chromosomes in mammals ­ contributes to critical aspects of physiology. Specifically, many of the metabolic traits impacted by myokines show striking differences arising from hormonal or genetic interactions depending on the genetic sex of the subject being studied. It is therefore important to consider genetic sex when studying the effects of myokines on the body. Velez, Van et al. wanted to gain a better understanding of how skeletal muscles interact with metabolic tissues such as pancreas, liver and brain, taking genetic sex into consideration. To do this they surveyed human datasets for the correlations between the activity of genes that code for myokines, the composition of muscle cells, the signaling between muscles and metabolic tissues and genetic sex. Their results showed that, genetic sex and sex hormones predicted most of the effects of skeletal muscle on other tissues. For example, myokines from muscle were predicted to be more impactful on liver or pancreas, depending on whether individuals were male or female, respectively. The results of Velez, Van et al. illustrate the importance of considering the effects of genetic sex and sexual hormones when studying metabolism. In the future, these results will allow other researchers to design sex-specific experiments to be able to gather more accurate information about the mechanisms of myokine signaling.


Cytokines , Receptors, Estradiol , Animals , Cytokines/metabolism , Female , Genetic Variation , Gonadal Steroid Hormones/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Receptors, Estradiol/metabolism
6.
Horm Mol Biol Clin Investig ; 42(3): 329-340, 2021 Mar 03.
Article En | MEDLINE | ID: mdl-34704691

The relationship between menopause and the development of metabolic diseases is well established. In postmenopause women, there is an expansion of visceral white adipose tissue (WATv), which highly contributes to the rise of circulating lipids. Meanwhile, muscle glucose uptake decreases and hepatic glucose production increases. Consequently, in the pancreas, lipotoxicity and glycotoxicity lead to deficient insulin production. These factors initiate an energy imbalance and enhance the probability of developing cardiovascular and metabolic diseases. Although the activation of estradiol receptors (ER) has been shown to be beneficial for the WAT stock pattern, leading to the insulin-sensitive phenotype, authors have described the risk of these receptors' activation, contributing to neoplasia development. The selective activation of beta-type ER (ERß) seems to be a promising strategy in the treatment of energy imbalance, acting on several tissues of metabolic importance and allowing an intervention with less risk for the development of estrogen-dependent neoplasia. However, the literature on the risks and benefits of selective ERß activation still needs to increase. In this review, several aspects related to ERß were considered, such as its physiological role in tissues of energy importance, beneficial effects, and risks of its stimulation during menopause. PubMed, SciELO, Cochrane, and Medline/Bireme databases were used in this study.


Biomarkers , Postmenopause/metabolism , Receptors, Estradiol/metabolism , Adipose Tissue/metabolism , Disease Susceptibility , Estrogens/metabolism , Female , Gene Expression Regulation , Humans , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Organ Specificity , Receptors, Estradiol/genetics , Receptors, Estrogen/metabolism , Signal Transduction
7.
Sci Rep ; 11(1): 7133, 2021 03 30.
Article En | MEDLINE | ID: mdl-33785829

Lactobacilli and estrogens play essential roles in vaginal homeostasis. We investigated the potential direct effect of 17ß-estradiol on a vaginal strain of Lactobacillus crispatus, the major bacterial species of the vaginal microbiota. 17ß-estradiol (10-6 to 10-10 M) had no effect on L. crispatus growth, but markedly affected the membrane dynamics of this bacterium. This effect appeared consistent with a signal transduction process. The surface polarity and aggregation potential of the bacterium were unaffected by exposure to 17ß-estradiol, but its mean size was significantly reduced. 17ß-estradiol also promoted biosurfactant production by L. crispatus and adhesion to vaginal VK2/E6E7 cells, but had little effect on bacterial biofilm formation activity. Bioinformatic analysis of L. crispatus identified a membrane lipid raft-associated stomatin/prohibitin/flotillin/HflK domain containing protein as a potential 17ß-estradiol binding site. Overall, our results reveal direct effects of 17ß-estradiol on L. crispatus. These effects are of potential importance in the physiology of the vaginal environment, through the promotion of lactobacillus adhesion to the mucosa and protection against pathogens.


Estradiol/physiology , Lactobacillus crispatus/physiology , Vagina/microbiology , Cell Adhesion , Cell Aggregation , Female , Humans , Membrane Fluidity , Receptors, Estradiol/metabolism
8.
Cell Mol Neurobiol ; 40(5): 711-723, 2020 Jul.
Article En | MEDLINE | ID: mdl-31784921

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.


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
9.
Cells ; 8(8)2019 08 19.
Article En | MEDLINE | ID: mdl-31430865

Oxidative stress has been considered the main mediator in neurodegenerative disease and in normal aging processes. Several studies have reported that the accumulation of reactive oxygen species (ROS), elevated oxidative stress, and neuroinflammation result in cellular malfunction. These conditions lead to neuronal cell death in aging-related neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease. Chronic administration of d-galactose (d-gal) for a period of 10 weeks causes ROS generation and neuroinflammation, ultimately leading to cognitive impairment. In this study, we evaluated the estrogen receptor α (ERα)/silent mating type information regulation 2 homolog 1 (SIRT1)-dependent antioxidant efficacy of 17ß-estradiol against d-gal-induced oxidative damage-mediated cognitive dysfunction in a male mouse model. The results indicate that 17ß-estradiol, by stimulating ERα/SIRT1, halts d-gal-induced oxidative stress-mediated JNK/NF-Ò¡B overexpression, neuroinflammation and neuronal apoptosis. Moreover, 17ß-estradiol ameliorated d-gal-induced AD-like pathophysiology, synaptic dysfunction and memory impairment in adult mouse brains. Interestingly, inhibition of SIRT1 with Ex527 (a potent and selective SIRT1 inhibitor) further enhanced d-gal-induced toxicity and abolished the beneficial effect of 17ß-estradiol. Most importantly, for the first time, our molecular docking study reveals that 17ß-estradiol allosterically increases the expression of SIRT1 and abolishes the inhibitory potential of d-ga. In summary, we can conclude that 17ß-estradiol, in an ERα/SIRT1-dependent manner, abrogates d-gal-induced oxidative stress-mediated memory impairment, neuroinflammation, and neurodegeneration in adult mice.


Cognitive Dysfunction/drug therapy , Estradiol/pharmacology , Neurodegenerative Diseases/drug therapy , Receptors, Estradiol/metabolism , Sirtuin 1/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Galactose , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/chemically induced , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
10.
Article En | MEDLINE | ID: mdl-31461684

Insulin-like growth factors (Igf1 and Igf2) play a key role in growth and development of vertebrates. In mammals, the expression of IGFs is regulated by estradiol-17ß (E2) via estrogen receptors (ESRs). The expression of igfs can also be regulated by E2 in fish, while comparative study of this is still lacking. The present study examined tissue distribution of igfs and hepatic expression of igfs and esrs during gonad development in Scatophagus argus by real-time PCR. Serum E2 concentration was measured by enzyme-linked immunosorbent assay (ELISA). The hepatic expression of igfs and esrs at gonadal phase III, incubated with either E2 (0.1, 1 or 10 µM) alone or in combination with estrogen receptor antagonists-fulvestrant, MPP or PHTPP, was measured. igf1 and igf2 expressed highest in liver of both sexes. Igf1, esr1 and esr2b expressions and serum E2 concentration increased, while igf2 and esr2a expressions decreased, during ovary development. Igfs and esrs expressions increased while serum E2 concentration maintained low during testis development. In females, E2 incubation enhanced the expressions of igf1 and esr1 but inhibited that of igf2 and esr2a. Both fulvestrant and MPP inhibited up-regulation effect of E2 on igf1 and esr1. Fulvestrant enhanced down-regulation effect of E2 on igf2 and esr2a, but MPP conversely. In males, E2 incubation enhanced the expressions of igfs, esr1 and esr2a. Fulvestrant and MPP inhibited up-regulation effect of E2 on igfs and esr1. PHTPP inhibited igf1 and esr2 expressions in both sexes. Our results indicated that the expression of igfs is regulated by E2 via Esrs in S. argus.


Estradiol/metabolism , Fishes/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Receptors, Estradiol/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Female , Fish Proteins/metabolism , Fishes/growth & development , Liver/growth & development , Liver/metabolism , Male , Ovary/growth & development , Ovary/metabolism , Testis/growth & development , Testis/metabolism
11.
Sci Rep ; 9(1): 9965, 2019 07 10.
Article En | MEDLINE | ID: mdl-31292456

The accessory ß1 subunit modulates the Ca2+- and voltage-activated K+ (BK) channel gating properties mainly by increasing its apparent Ca2+ sensitivity. ß1 plays an important role in the modulation of arterial tone and blood pressure by vascular smooth muscle cells (SMCs). 17ß-estradiol (E2) increases the BK channel open probability (Po) in SMCs, through a ß1 subunit-dependent modulatory effect. Here, using molecular modeling, bioinformatics, mutagenesis, and electrophysiology, we identify a cluster of hydrophobic residues in the second transmembrane domain of the ß1 subunit, including the residues W163 and F166, as the binding site for E2. We further show that the increase in Po induced by E2 is associated with a stabilization of the voltage sensor in its active configuration and an increase in the coupling between the voltage sensor activation and pore opening. Since ß1 is a key molecular player in vasoregulation, the findings reported here are of importance in the design of novel drugs able to modulate BK channels.


Estradiol/metabolism , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Myocytes, Smooth Muscle/metabolism , Calcium/metabolism , HEK293 Cells , Humans , Membrane Potentials , Patch-Clamp Techniques/methods , Protein Subunits , Receptors, Estradiol/metabolism
12.
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
13.
Steroids ; 142: 34-42, 2019 02.
Article En | MEDLINE | ID: mdl-29274403

There is growing evidence that laryngeal cancers are responsive to sex hormones, specifically 17ß-estradiol (E2), despite controversy regarding the presence and characterization of E2 receptors (ER). Determination of sex hormone responsiveness impacts the prognosis of laryngeal cancer patients and the treatment modalities implemented by their clinicians. Discovery of membrane-associated steroid hormone receptors and rapid membrane signaling opened the possibility that cancers previously labeled 'non-hormone dependent' and 'ER negative' might in fact be susceptible to the effects of E2 via these membrane receptors. ERα66 and ERß, the classical nuclear receptors, are present in the membranes of different cancer cells via a mechanism referred to as trafficking. Novel splice variants of these traditional receptors, a key example being ERα36, have also been found in the caveolae of cancer cells. Previous work demonstrated that ERα36 has a role in the tumorigenesis of laryngeal cancer, enhancing both proliferation and the anti-apoptotic effect of E2 against chemotherapeutics. The present study showed that expression of different membrane ERs in laryngeal cancer is not uniform, which may result in differential and even antagonistic responses to E2. E2 had protective or deleterious effects in different cancer cell lines, stimulating proliferation and conferring anti-apoptotic potential to the cancer cells according to their receptor profile. These findings stress the importance of establishing the molecular and clinical characterization of the specific laryngeal tumor in order to tailor treatment accordingly, thus optimizing care while reducing adverse effects for individual patients.


Estradiol/pharmacology , Estrogens/metabolism , Estrogens/pharmacology , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/metabolism , Receptors, Estradiol/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Estradiol/metabolism , Humans , Laryngeal Neoplasms/pathology , Receptors, Estradiol/analysis , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Horm Behav ; 107: 20-25, 2019 01.
Article En | MEDLINE | ID: mdl-30462987

Activation of membrane-associated estrogen receptors (mER) decreases food and water intake in female rats. Additional studies suggest these effects are mediated, at least in part, by membrane-associated estrogen receptor alpha (ERα). Nevertheless, the critical site of action and the intracellular signaling required for the ingestive effects of ERα remain unclear. Estradiol given to the medial preoptic area (mPOA) decreases ingestive behaviors, and membrane-associated ERα has been shown to affect intracellular signaling through interactions with metabotropic glutamate receptor (mGluR) subtypes, but an involvement of this signaling pathway, in the mPOA, in ingestive behavior remains untested. To address these open questions, we first showed that activation of mER in the mPOA decreased both overnight food and water intake, and did so in a time course consistent with a genomic mechanism of action. Next, we tested the requirement of mGluR1a signaling in the mPOA for the anorexigenic and anti-dipsogenic effects of estradiol. As expected, estradiol in the mPOA decreased food intake, but only in the absence of an mGluR1a antagonist. The same was not true for estradiol effects on water intake, which were unaffected by an mGluR1a antagonist. These results suggest that estrogens require mGluR activation for at least some of their effects on ingestive behaviors, and indicate that the mPOA is a critical site of action. The results also reveal an interesting divergence in the estrogenic control of ingestive behavior by which mGluR signaling in the mPOA plays a role in the control of food intake, but not water intake.


Anorexia/chemically induced , Appetite Depressants/pharmacology , Estradiol/pharmacology , Preoptic Area/drug effects , Receptors, Estradiol/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Anorexia/metabolism , Drinking/drug effects , Eating/drug effects , Female , Preoptic Area/metabolism , Rats , Rats, Long-Evans , Signal Transduction/drug effects
15.
Acta Physiol (Oxf) ; 222(4): e13001, 2018 04.
Article En | MEDLINE | ID: mdl-29178430

AIM: Pregnant women have been recommended to take FA daily to prevent birth defects in the brain and spinal cord. We previously showed that folic acid (FA) exerts an anti-angiogenic activity. As angiogenesis is important for endometrial reorganization and embryonic development, there should be some mechanisms to allow the pregnant mother and the foetus to escape from the FA-induced anti-angiogenesis. This study was designed to investigate the effect of female sex hormones on the FA-induced anti-angiogenic activity. METHODS: The protein levels and protein-protein interaction were examined by Western blot analysis and immunoprecipitation assay respectively. The cell proliferation and migration were examined by MTT assay and wound healing assay respectively. The in vivo angiogenesis was evaluated by Matrigel angiogenesis assay. RESULTS: In human umbilical venous endothelial cells (HUVEC), FA receptor (FR) formed a complex with progesterone receptor (PR), oestradiol receptor (ER) and cSrc. Pregnancy levels of progesterone (P4) or oestradiol (E2) prevented FA-induced inhibitions of proliferation and migration in HUVEC. Both E2 and P4 prevented the FA-induced anti-angiogenesis in vivo. Moreover, cotreatment with FA and P4 or E2 inhibited the signalling pathways involved in FA-induced inhibitions of proliferation and migration in HUVEC. CONCLUSION: Female sex hormones interrupt the FA-induced anti-angiogenic action through receptor-receptor interaction.


Angiogenesis Inhibitors/pharmacology , Estradiol/pharmacology , Folic Acid/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Progesterone/pharmacology , Animals , Cell Proliferation/drug effects , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Pregnancy , Receptors, Estradiol/metabolism , Receptors, Progesterone/metabolism
16.
Sci Rep ; 7(1): 139, 2017 03 10.
Article En | MEDLINE | ID: mdl-28273940

Previously, we demonstrated that chronic exposure to low levels of estradiol-17ß (E2) increases mean arterial pressure (MAP) in young female Sprague-Dawley (SD) rats, however, the underlying mechanisms are unclear. Since endothelin-1 (ET-1) is implicated in blood pressure (BP) regulation, we hypothesized that E2's effects on MAP are mediated through central ET-1. To test this, young female SD rats were either sham implanted or implanted s.c. with slow-release E2 pellets (20 ng/day for 90 days). BP was monitored by telemetry. After 75 days of E2 exposure, ETA antagonist or vehicle was administered i.c.v. After 90 days of E2 exposure, rats were sacrificed, and the paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM) were microdissected for gene expression and protein analysis of ET-1 and its receptors. E2 exposure increased MAP after pellet implantation. Gene expression of ET-1 and ETA but not ETB receptors were upregulated in the PVN and RVLM of E2 treated animals. Further, the protein levels of ETA receptor were also increased in the PVN of E2 treated animals. However, i.c.v. infusion of the ETA antagonist did not completely block the increase in blood pressure. Our results suggest that increases in central ET-1 activity could possibly play a role in chronic E2-induced increase in BP but further studies are needed to completely understand the contribution of ET-1 in this phenomenon.


Endothelin-1/genetics , Endothelin-1/metabolism , Estradiol/toxicity , Estrogen Antagonists/administration & dosage , Hypertension/chemically induced , Animals , Blood Pressure/drug effects , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hypertension/genetics , Hypertension/metabolism , Medulla Oblongata/chemistry , Medulla Oblongata/drug effects , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Estradiol/genetics , Receptors, Estradiol/metabolism , Toxicity Tests, Chronic
17.
J Morphol ; 277(4): 412-23, 2016 Apr.
Article En | MEDLINE | ID: mdl-26719144

The dynamic equilibrium between spermatogonial proliferation and testicular apoptosis determines the progression of spermatogenesis in amphibians. Estrogens and their receptors play a central role in regulating spermatogenesis in vertebrates, and in some species of anurans, estradiol (E2 ) is involved in the regulation of spermatogonial proliferation and apoptosis of germ cells. Bidder's organ (BO) is a structure characteristic of Bufonidae that has historically been compared to an undeveloped ovary. In adult Rhinella arenarum males, BO is one of the main sources of plasma E2 . The aim of this study was 1) to describe the seasonal variations in testicular apoptosis, spermatogonial proliferation, and cellular proliferation in BO; and 2) to analyze the presence and localization of estrogen receptor ß (ERß) in the testes and BO of R. arenarum. Testicular fragments and BOs from animals collected during the year were labeled with 5-bromo-2'-deoxyuridine (BrdU) and BrdU incorporation was determined using immunohistochemistry. Apoptosis in testicular sections was detected using the TUNEL method, and ERß localization was assessed using immunohistochemistry in testes and BOs. The results indicate that spermatogonial proliferation is highest during the reproductive season and that cysts of spermatocytes and spermatids undergo apoptosis during the postreproductive season. Furthermore, the proliferation of follicular cells is highest during the reproductive and postreproductive seasons. ERß was primarily detected by immunolocalization in Sertoli cells, follicular cells, and oocytes. Taken together, these results suggest that cysts that do not form spermatozoa are removed from testes by apoptosis and that estrogens regulate both spermatogenesis and oogenesis in adult males of R. arenarum.


Apoptosis/physiology , Bufonidae/physiology , Cell Proliferation , Spermatogenesis/physiology , Testis/cytology , Testis/physiology , Animals , Bufonidae/anatomy & histology , Estrogen Receptor beta/metabolism , Male , Receptors, Estradiol/metabolism , Sertoli Cells
18.
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
19.
Compr Physiol ; 5(3): 1211-22, 2015 Jul 01.
Article En | MEDLINE | ID: mdl-26140715

The discoveries of rapid, membrane-initiated steroid actions and central nervous system steroidogenesis have changed our understanding of the neuroendocrinology of reproduction. Classical nuclear actions of estradiol and progesterone steroids affecting transcription are essential. However, with the discoveries of membrane-associated steroid receptors, it is becoming clear that estradiol and progesterone have neurotransmitter-like actions activating intracellular events. Ultimately, membrane-initiated actions can influence transcription. Estradiol membrane-initiated signaling (EMS) modulates female sexual receptivity and estrogen feedback regulating the luteinizing hormone (LH) surge. In the arcuate nucleus, EMS activates a lordosis-regulating circuit that extends to the medial preoptic nucleus and subsequently to the ventromedial nucleus (VMH)--the output from the limbic and hypothalamic regions. Here, we discuss how EMS leads to an active inhibition of lordosis behavior. To stimulate ovulation, EMS facilitates astrocyte synthesis of progesterone (neuroP) in the hypothalamus. Regulation of GnRH release driving the LH surge is dependent on estradiol-sensitive kisspeptin (Kiss1) expression in the rostral periventricular nucleus of the third ventricle (RP3V). NeuroP activation of the LH surge depends on Kiss1, but the specifics of signaling have not been well elucidated. RP3V Kiss1 neurons appear to integrate estradiol and progesterone information which feeds back onto GnRH neurons to stimulate the LH surge. In a second population of Kiss1 neurons, estradiol suppresses the surge but maintains tonic LH release, another critical component of the estrous cycle. Together, evidence suggests that regulation of reproduction involves membrane action of steroids, some of which are synthesized in the brain.


Gonadal Steroid Hormones/metabolism , Receptors, Estradiol/metabolism , Reproduction , Signal Transduction , Animals , Brain/metabolism , Female , Gonadotropins, Pituitary/metabolism , Humans , Kisspeptins/metabolism
20.
Cancer Radiother ; 19(5): 295-302, 2015 Aug.
Article En | MEDLINE | ID: mdl-26188735

PURPOSE: Several studies showed a breast cancer downstaging due to screening. A first national survey was conducted in France in 2001-2002 to evaluate in the current clinical practice the clinicopathological features and treatments of 1049 firstly operated breast cancers. In order to assess the impact of the national screening program implemented in all regions in France in 2004, a new survey was performed in 2007-2008. MATERIAL: The new survey included 1433 firstly operated breast cancers prospectively collected. These new data were compared to the results of the first national survey. RESULTS: According to TN classification, we found in the second survey T0: 27.6%, T1: 48.6%, T2: 21.3%, T3T4: 3.8% and Tx: 0.7%. Infiltrating ductal and lobular carcinomas represented 80% and 13% of tumours. Hormone receptors were positive in 85.3% and Her-2 overexpressed in 12.4% of tumours (83.9% and 20.6% in the first survey); 68.2% and 32% were pN0 and pN1-3. Lumpectomy and mastectomy were performed in 77% and 23% of the cases. Axillary dissection, sentinel node biopsy or both were performed in 42.6%, 41% and 16.4% of the cases, respectively. Radiotherapy, chemotherapy, hormonotherapy and trastuzumab were given to 93%, 51%, 83% and 9.3% of the patients. Compared with the results from the first survey, we found an increase of infraclinical lesions (T0 from 8.4 to 27.6%) and a wide decrease of pN+ rate (from 44% to 32%). The mastectomy rate was constant (23%), as well as radiotherapy use, whereas chemotherapy use decreased from 62.8 to 55.6%. CONCLUSION: A complete national screening coverage clearly provides a favourable modification of breast cancer clinicopathological features. Both locoregional and adjuvant treatments were greatly downscaled.


Breast Neoplasms/pathology , Breast Neoplasms/therapy , Mass Screening , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/epidemiology , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/epidemiology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/epidemiology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Carcinoma, Lobular/therapy , Chemotherapy, Adjuvant/statistics & numerical data , Female , France/epidemiology , Health Surveys , Humans , Lymph Node Excision/statistics & numerical data , Mastectomy/statistics & numerical data , Mastectomy, Segmental/statistics & numerical data , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prospective Studies , Radiotherapy, Adjuvant , Receptor, ErbB-2/metabolism , Receptors, Estradiol/metabolism , Receptors, Progesterone/metabolism , Sentinel Lymph Node Biopsy/statistics & numerical data , Trastuzumab , Young Adult
...