Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Obes (Lond) ; 41(6): 945-954, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28220039

RESUMEN

BACKGROUND: Treatment with estrogen in early menopausal women protects against development of hepatic steatosis and nonalcoholic fatty liver disease but estrogen has undesirable side effects, which negate its beneficial effects in premenopausal and postmenopausal women. Targeted therapies require better understanding of the target sites and mechanisms by which estrogen signaling exerts its protective effects in women. Estrogen receptor α (ERα) is thought to be the primary mediator for estrogen signaling to protect against hepatic steatosis. ERα has several mechanisms for signal transduction: (1) inducing gene transcription by direct binding to specific DNA sequences, (2) inducing tethered transcription with other DNA-binding factors, and (3) stimulating nongenomic action through membrane-associated ERα. However, it is still unclear which mechanisms mediate ERα-dependent protection against hepatic steatosis. METHODS: To understand the mechanisms of estrogen signaling for protection against hepatic steatosis in females, we analyzed the global ERα knockout mouse (αERKO), ERα DNA-binding domain mutant mouse (KIKO) and liver-specific ERα knockout mouse (LERKO) fed high-fat diets (HFD). The KIKO mouse disrupts the direct DNA-binding transcription activity but retains tethered transcription regulation and nongenomic action. Hepatic steatosis was evaluated by scoring the macrovesicular and microvesicular steatosis as well as serum alanine aminotransferase (ALT) levels. We analyzed serum testosterone to assess its correlation with hepatic steatosis. RESULTS: Liver fat accumulation was far greater in HFD-fed αERKO and KIKO females than in HFD-fed wild-type (WT) controls. Conversely, HFD-fed LERKO females did not accumulate excess liver fat. HFD-fed αERKO and KIKO females showed higher microvesicular steatosis and ALT levels than WT controls that correlated with increased serum testosterone levels. CONCLUSIONS: ERα-mediated direct transcription in non-hepatic tissues is essential for estrogen-mediated protection against hepatic steatosis in HFD-fed females. The balance between non-hepatic estrogen signaling and hepatic or non-hepatic testosterone action may control hepatic steatosis.


Asunto(s)
Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Adiposidad , Animales , Western Blotting , Proteínas de Unión al ADN/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Estrógenos/administración & dosificación , Femenino , Ratones , Ratones Noqueados , Factores de Transcripción/efectos de los fármacos
2.
Handb Exp Pharmacol ; (198): 29-35, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20839084

RESUMEN

Estrogens influence fertility and infertility in animals. This chapter reviews the use of estrogen as a contraceptive through the regulation of its production and action. It is concluded that the use of specific agonists and antagonists of estrogen action that avoid the global and unwanted side effects of estrogen offers new potential methods of contraception.


Asunto(s)
Estrógenos/fisiología , Reproducción/fisiología , Transducción de Señal/fisiología , Animales , Anticonceptivos Hormonales Orales/efectos adversos , Anticonceptivos Hormonales Orales/farmacología , Estrógenos/biosíntesis , Femenino , Humanos
3.
Genes Immun ; 9(2): 137-52, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18200028

RESUMEN

Estrogens promote lupus in humans and some mouse models of this disease. Nonetheless, little is known about the role of estrogen receptors in lupus pathogenesis. Here, we report that in females on the lupus-prone (NZB x NZW)F(1) background, disruption of estrogen receptor-alpha (ER alpha or Esr1) attenuated glomerulonephritis and increased survival. ER alpha deficiency also retarded development of anti-histone/DNA antibodies, suggesting that ER alpha promotes loss of immunologic tolerance. Furthermore, ER alpha deficiency in (NZB x NZW)F(1) females attenuated the subsequent development of anti-double-stranded DNA (dsDNA) IgG antibodies, which are associated with glomerulonephritis in this model. We provide evidence that ER alpha may promote lupus, at least in part, by inducing interferon-gamma, an estrogen-regulated cytokine that impacts this disease. ER alpha deficiency in (NZB x NZW)F(1) males increased survival and reduced anti-dsDNA antibodies, suggesting that ER alpha also modulates lupus in males. These studies demonstrate that ER alpha, rather than ER beta, plays a major role in regulating autoimmunity in (NZB x NZW)F(1) mice. Furthermore, our results suggest for the first time that ER alpha promotes lupus, at least in part, by impacting the initial loss of tolerance. These data suggest that targeted therapy disrupting ER alpha, most likely within the immune system, may be effective in the prevention and/or treatment of lupus.


Asunto(s)
Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Animales , Receptor alfa de Estrógeno/fisiología , Femenino , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Endogamia , Lupus Eritematoso Sistémico/prevención & control , Masculino , Ratones , Ratones Endogámicos NZB , Ratones Noqueados
4.
Genes Brain Behav ; 5(7): 528-39, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17010099

RESUMEN

Social recognition, processing, and retaining information about conspecific individuals is crucial for the development of normal social relationships. The neuropeptide oxytocin (OT) is necessary for social recognition in male and female mice, with its effects being modulated by estrogens in females. In previous studies, mice whose genes for the estrogen receptor-alpha (alpha-ERKO) and estrogen receptor-beta (beta-ERKO) as well as OTKO were knocked out failed to habituate to a repeatedly presented conspecific and to dishabituate when the familiar mouse is replaced by a novel animal (Choleris et al. 2003, Proc Natl Acad Sci USA 100, 6192-6197). However, a binary social discrimination assay, where animals are given a simultaneous choice between a familiar and a previously unknown individual, offers a more direct test of social recognition. Here, we used alpha-ERKO, beta-ERKO, and OTKO female mice in the binary social discrimination paradigm. Differently from their wild-type controls, when given a choice, the KO mice showed either reduced (beta-ERKO) or completely impaired (OTKO and alpha-ERKO) social discrimination. Detailed behavioral analyses indicate that all of the KO mice have reduced anxiety-related stretched approaches to the social stimulus with no overall impairment in horizontal and vertical activity, non-social investigation, and various other behaviors such as, self-grooming, digging, and inactivity. Therefore, the OT, ER-alpha, and ER-beta genes are necessary, to different degrees, for social discrimination and, thus, for the modulation of social behavior (e.g. aggression, affiliation).


Asunto(s)
Discriminación en Psicología/fisiología , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Oxitocina/fisiología , Conducta Social , Análisis de Varianza , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Conducta Exploratoria/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Actividad Motora/fisiología , Análisis Multivariante , Oxitocina/genética
5.
Mol Carcinog ; 45(7): 490-505, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16482517

RESUMEN

The inappropriate activation of one or more members of the ErbB family of receptor tyrosine kinases [ErbB-1 (EGFR), ErbB-2, ErbB-3, ErbB-4] has been linked with oncogenesis. ErbB-2 is frequently coexpressed with ErbB-3 in breast cancer cells and in the presence of the ligand heregulin (HRG) the ErbB-2/ErbB-3 receptors form a signaling heterodimer that can affect cell proliferation and apoptosis. The major goal of the present study was to determine whether endogenous HRG causes autocrine/paracrine activation of ErbB-2/ErbB-3 and contributes to the proliferation of mammary epithelial tumor cells. Tyrosine-phosphorylated (activated) ErbB-2 and ErbB-3 receptors were detected in the majority of extracts from tumors that had formed spontaneously or as a result of oncogene expression. HRG-1 transcripts and protein were found in the epithelial cells of most of these mouse mammary tumors. Various mouse mammary cell lines also contained activated ErbB-2/ErbB-3 and HRG transcripts. A approximately 50 kDa C-terminal fragment of pro-HRG was detected, which indicates that the HRG-1 precursor is readily processed by these cells. It is likely that the secreted mature HRG activated the ErbB-2/3 receptors. Addition of an antiserum against HRG to the mammary epithelial tumor cell line TM-6 reduced ErbB-3 Tyr-phosphorylation. Treatment with HRG-1 siRNA oligonucleotides or infection with a retroviral construct to stably express HRG siRNA effectively reduced HRG protein levels, ErbB-2/ErbB-3 activation, and the rate of proliferation, which could be reversed by the addition of HRG. The cumulative findings from these experiments show that coexpression of the HRG ligand contributes to activation of ErbB-2/Erb-3 in mouse mammary tumor cells in an autocrine or paracrine fashion.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Neurregulina-1/genética , Receptor ErbB-2/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Cartilla de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Neurregulina-1/metabolismo , Fragmentos de Péptidos/química , Plásmidos , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Receptor ErbB-2/genética , Proteínas Recombinantes/metabolismo , Transfección
6.
Endocrinology ; 147(1): 191-200, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16223864

RESUMEN

Estrogens induce both proliferative and antiproliferative responses in the prostate gland. To date, antiproliferative effects of estrogens are generally considered to be due to systemic antiandrogenic actions. However, estrogen action mediated through estrogen receptor (ER) beta was recently suggested as another mechanism of induction of apoptosis in the prostate. This study aimed to explore the hypothesis that the antiproliferative effects of estrogen are directly mediated through ERbeta using a prostate organ culture system. We previously reported effects of 17beta-estradiol (E2) using rat ventral prostate (VP) tissues, and adapted the system for culturing mouse tissues. In both rat and mouse models, estrogen-induced apoptosis was detected that was spatially and regionally localized to the epithelium of the distal tips. Using organ cultures of alphaER knockout (alphaERKO) and betaERKO prostates, we failed to demonstrate that apoptosis induced by E2 was mediated through either receptor subtype. Activation of ER-selective ligands (ERalpha, propyl pyrazole triol, ERbeta, diaryl-proprionitrile, and 5alpha-androstane-3beta,17beta-diol) in organ culture experiments failed to induce apoptosis, as did the membrane impermeable conjugate E2:BSA, discounting the possibility of nongenomic effects. Consequently, E2 regulation of androgen receptor (AR) expression was examined and, in the presence of nanomolar testosterone levels, E2 caused a specific reduction in AR protein expression in wild-type, alphaERKO, and betaERKO mice, particularly in the distal region where apoptosis was detected. This down-regulation of AR protein provides a possible mechanism for the proapoptotic action of E2 that is independent of ERs or nongenomic effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Próstata/citología , Próstata/fisiología , Animales , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/genética , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Próstata/efectos de los fármacos , Ratas
7.
Artículo en Inglés | MEDLINE | ID: mdl-17824169

RESUMEN

Hormonal effects on classical endocrine target organs such as the female reproductive tract, mammary gland, ovary, and neuroendocrine system have been thoroughly studied, with significant advancements in our understanding of estrogen actions and disease conditions from both cell culture as well as new experimental animal models. Knowledge of the highly appreciated effects of estrogen in nonclassical endocrine organ systems, arising from epidemiological and clinical findings in the cardiovascular, immune, GI tract, and liver, is only now becoming clarified from the development and use of knock-out or transgenic animal models for the study of both estrogen and ER activities. There are considerable epidemiological data showing that premenopausal females (Barrett-Connor 1997; Crabbe et al. 2003) have reduced risk for cardiovascular disease. However, a recent large clinical trial failed to show cardioprotection for postmenopausal females on estrogen-progestin replacement (Rossouw et al. 2002). In fact, the Women's Health Initiative Study showed increased cardiovascular risk for females taking an estrogen-progestin combination. These studies suggest that we need a better understanding of the mechanisms responsible for cardioprotection in females.


Asunto(s)
Cardiomegalia/etiología , Estrógenos/fisiología , Daño por Reperfusión Miocárdica/etiología , Receptores de Estrógenos/fisiología , Animales , Femenino , Humanos , Masculino , Caracteres Sexuales
8.
Neuroscience ; 130(2): 445-56, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15664701

RESUMEN

We examined the distribution of estrogen receptor (ER)-alpha and ER-beta immunoreactive (ir) cells in the dorsal (DRN) and median/paramedian (MPRN) raphe nuclei in male mice. ER-alpha ir neurons were scattered across the three subdivisions (ventral, dorsal, and lateral) of the DRN and the MPRN. Robust ER-beta ir cells were observed throughout the raphe nuclei, and were particularly abundant in the ventral and dorsal subdivisions of the DRN. Using dual-label immunocytochemistry for ER-alpha or ER-beta with tryptophan hydroxylase (TPH), the rate-limiting enzyme for 5-hydroxytryptamine (5-HT) synthesis, over 90% of ER-beta ir cells exhibited TPH-ir in all DRN subdivisions, whereas only 23% of ER-alpha ir cells contained TPH. Comparisons of ER-alpha knockout (alphaERKO) as well as ER-beta knockout (betaERKO) mice with their respective wild-type (WT) littermates revealed that gene disruption of either ER-alpha or ER-beta did not affect the other ER subtype expression in the raphe nuclei. In situ hybridization histochemistry revealed that there was a small but statistically significant decrease in TPH mRNA expression in the ventral DRN subdivision in betaERKO mice compared with betaWT mice, whereas TPH mRNA levels were not affected in alphaERKO mice. These findings support a hypothesis that ER-beta activation may contribute to the estrogenic regulation of neuroendocrine and behavioral functions, in part, by acting directly on 5-HT neurons in the raphe nuclei in male mice.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Vías Nerviosas/metabolismo , Sustancia Gris Periacueductal/metabolismo , Núcleos del Rafe/metabolismo , Serotonina/metabolismo , Animales , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Vías Nerviosas/anatomía & histología , Neuronas/metabolismo , Sustancia Gris Periacueductal/anatomía & histología , ARN Mensajero/metabolismo , Núcleos del Rafe/anatomía & histología , Factores Sexuales , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
9.
Genes Brain Behav ; 3(4): 189-95, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15307217

RESUMEN

Social behavior involves both the recognition and pro-duction of social cues. Mice with selective deletion(knockout) of either the gene for oxytocin (OT) or genes for the estrogen receptor (ER) -c or -B display impaired social recognition. In this study we demonstrate that these gene knockout mice also provide discriminably different social stimuli in behavioral assays. In an odor choice test, which is a measure of social interest and discrimination, outbred female Swiss-Webster mice discriminated the urine odors of male knock-outs IKO: OTKO, alphaERKO, betaERKO) from the odors of their wildtype littermates (WT: OTWT, alphaERWT, betaERWT). Females showed marked initial choices of the urine odors of OTWT and betaERWT males over those of OTKOand PERKO males, and alphaERKO males over alphaERWT males. The odors of OTKO and betaERKO males also induced aversive, analgesic responses, with the odors of WTs having no significant effects. Odors of both the alphaERWT andalphaERKO males induced aversive, analgesic responses,with the odors of the WT inducing significantly greater analgesia. The odors of restraint stressed WT and KO males also elicited analgesia with, again, females dis-playing significantly greater responses to the odors of stressed OTKO and betaERKO males than their WTs, and significantly lower analgesia to the odors of stressedalphaERKO than alphaERWT males. These findings show that the KO mice are discriminated from their WTs on the basis of odor and that the various KOs differ in the relative attractiveness/aversiveness of their odors. Therefore, in behavioral assays one causal route by which gene inactivation alters the social behavior of knockout mice may be mediated through the partners'modified responses to their odors.


Asunto(s)
Conducta Animal/fisiología , Oxitocina/genética , Feromonas/genética , Feromonas/orina , Receptores de Estrógenos/genética , Conducta Social , Animales , Discriminación en Psicología/fisiología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Reconocimiento en Psicología/fisiología
10.
Biol Reprod ; 71(1): 273-81, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15028626

RESUMEN

Transformation of the endometrium into the receptive phase is under the control of ovarian steroid hormones and is modulated by embryonic signals during implantation. We have previously shown that this differentiation process is accompanied by a suppression of gap junction connexins (Cx) 26 and 43 before implantation followed by a local induction of both connexins in the implantation chamber. In the present study, we demonstrate that connexin gene expression in the rodent endometrium is regulated via two distinct signaling pathways during these different stages of early pregnancy. During preimplantation, transcription of connexins can be induced by estrogen via an estrogen receptor (ER)-dependent pathway. Additionally, Cx26 and Cx43 are induced by embryonic signals during implantation and delayed implantation as well as during artificially induced decidualization. In contrast to the estrogen-induced expression, this embryonic/decidual-associated induction of Cx26 and Cx43 could not be blocked by antiestrogen, thus pointing to another regulatory pathway independent of the ER. Studies in ERalpha and ERbeta knockout mice confirmed these different pathways, demonstrating that in the endometrium, estrogen-mediated Cx26 gene induction, but not induction during decidualization, is dependent on functional ERalpha. To evaluate potential embryonic signals regulating Cx26 expression, uteri of pseudopregnant animals were incubated with different mediators in an organ-culture model, showing that catechol estrogen and mediators of the inflammatory cascade such as prostaglandin F(2alpha) and interleukin-1beta are able to induce Cx26 expression through the ER-independent pathway. Thus, the present study demonstrates that endometrial expression of Cx26 and Cx43 is induced via estrogen and ERalpha during preimplantation but then utilizes an ER-independent signaling pathway during embryo implantation and decidualization.


Asunto(s)
Conexina 43/metabolismo , Conexinas/metabolismo , Implantación del Embrión/fisiología , Endometrio/metabolismo , Estrógenos/fisiología , Animales , Conexina 26 , Conexina 43/genética , Conexinas/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/fisiología , Femenino , Expresión Génica , Regulación de la Expresión Génica , Ratones , Ratones Noqueados/genética , Embarazo , Ratas , Ratas Sprague-Dawley , Activación Transcripcional
11.
J Steroid Biochem Mol Biol ; 86(3-5): 477-86, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14623547

RESUMEN

Long term exposure to estradiol increases the risk of breast cancer in a variety of animal species, as well as in women. The mechanisms responsible for this effect have not been firmly established. The prevailing theory proposes that estrogens increase the rate of cell proliferation by stimulating estrogen receptor-mediated transcription and thereby the number of errors occurring during DNA replication. An alternative hypothesis proposes that estradiol can be metabolized to quinone derivatives which can react with DNA and then remove bases from DNA through a process called depurination. Error prone DNA repair then results in point mutations. We postulate that these two processes, increased cell proliferation and genotoxic metabolite formation, act in an additive or synergistic fashion to induce cancer. If correct, aromatase inhibitors would block both processes whereas anti-estrogens would only inhibit receptor-mediated effects. Accordingly, aromatase inhibitors would be more effective in preventing breast cancer than use of anti-estrogens. Our studies initially demonstrated that catechol estrogen (CE) quinone metabolites are formed in MCF-7 human breast cancer cells in culture. Measurement of estrogen metabolites and conjugates involved utilization of an HPLC separation coupled with an electrochemical detector. We then utilized an animal model that allows dissociation of estrogen receptor-mediated function from that of the effects of estradiol metabolites. Wnt-1 transgenic mice harboring a knock-out of ERalpha provides a means of examining the effect of estrogen deprivation in the absence of the ER in animals with a high incidence of breast tumors. ERbeta was shown to be absent in the breast tissue of these animals by RNase protection assay. In the breast tissue of these estrogen receptor alpha knock-out (ERKO)/Wnt-1 transgenic mice, we demonstrated formation of genotoxic estradiol metabolites. The ERKO/Wnt-1 breast extracts contained picomole amounts of the 4-catechol estrogens, but not their methoxy conjugates nor the 2-CE or their methoxy conjugates. The 4-CE conjugates with glutathione or its hydrolytic products (cysteine and N-acetylcysteine) were detected in picomole amounts in both tumors and hyperplastic mammary tissue, demonstrating the formation of CE-3,4-quinones. These results are consistent with the hypothesis that mammary tumor development is primarily initiated by metabolism of estrogens to 4-CE and, then, to CE-3,4-quinones, which may react with DNA to induce oncogenic mutations. The next set of experiments examined the incidence of tumors formed in Wnt-1 transgenic mice bearing wild type ERalpha (ER+/+), the heterozygous combination of genes (ER+/ER-) or ERalpha knock-out (ER-/-). To assess the effect of estrogens in the absence of ER, half of the animals were oophorectomized on day 15 and the other half were sham operated. Castration reduced the incidence of breast tumors in all animal groups and demonstrated the dependence of tumor formation upon estrogens. A trend toward reduction in tumor number (not statistically significant at this interim analysis) occurred in the absence of functional ER since the number of tumors was markedly reduced in ERKO animals which were castrated early in life. In aggregate, our results support the concept that metabolites of estradiol may act in concert with ER mediated mechanisms to induce breast cancer.


Asunto(s)
Neoplasias de la Mama/inducido químicamente , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Estradiol/metabolismo , Estradiol/toxicidad , Neoplasias Mamarias Animales/inducido químicamente , Animales , Aromatasa/genética , Aromatasa/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Catecol O-Metiltransferasa/genética , División Celular/genética , Estrona/análogos & derivados , Estrona/metabolismo , Humanos , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Mutación , Polimorfismo Genético , Factores de Riesgo
12.
Proc Natl Acad Sci U S A ; 100(19): 11019-22, 2003 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12930901

RESUMEN

We have identified a generalized arousal component in the behavior of mice. Analyzed by mathematical/statistical approaches across experiments, investigators, and mouse populations, it accounts for about 1/3 of the variance in arousal-related measures. Knockout of the gene coding for the classical estrogen receptor (ER-alpha), a ligand-activated transcription factor, greatly reduced arousal responses. In contrast, disrupting the gene for a likely gene duplication product, ER-beta, did not have these effects. A combination of mathematical and genetic approaches to arousal in an experimentally tractable mammal opens up analysis of a CNS function of considerable theoretical and practical significance.


Asunto(s)
Encéfalo/fisiología , Genética , Conducta Sexual Animal , Animales , Ratones
13.
Gynecol Endocrinol ; 17(2): 169-76, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12737678

RESUMEN

There is a broad spectrum of organ systems that respond to estrogen hormones, including the female and male reproductive tracts, mammary gland, the skeleton, cardiovascular system and central nervous system. The physiological effects of estrogens are mediated by the estrogen receptor, a member of the nuclear receptor superfamily of transcription factors. Two estrogen receptors have been identified: the originally described estrogen receptor alpha (ERalpha) and the more recently discovered estrogen receptor beta (ERbeta). Three different estrogen receptor knockout (ERKO) mouse models were generated, carrying a null mutation in the ERalpha gene (alphaERKO), the ERbeta gene (betaERKO) or both genes (alphabetaERKO). The generation of the different ERKO mice provides ideal models for studying the physiological consequences of the complete lack of estrogen receptor activity and the distinct roles of both estrogen receptors in various tissues. This review summarizes the phenotypes principally seen in the female reproductive system of the different ERKO mice.


Asunto(s)
Genitales Femeninos/química , Genitales Femeninos/fisiología , Receptores de Estrógenos/deficiencia , Animales , Resistencia a Medicamentos/genética , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Estrógenos/farmacología , Femenino , Humanos , Ratones , Ratones Noqueados , Mutación , Ovario/química , Ovario/fisiología , Fenotipo , Receptores de Estrógenos/genética , Receptores de Estrógenos/fisiología , Reproducción , Útero/química , Útero/fisiología
14.
Placenta ; 24(4): 403-11, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12657515

RESUMEN

Uterine Natural Killer (uNK) cell differentiation in vivo requires oestrogen (E) priming prior to progesterone (P). Hybridomas between uNK precursor and SP2/0 cells express message for E receptor (ER)alpha but nor PR. However, mature, rodent and human uNK cells lack these receptors. To functionally assess requirements for uNK cell expression of ERalpha or ERbeta during precursor differentiation, marrow was transplanted from either ERalpha(o/o) (alphaERKO) or ERbeta(o/o) (betaERKO) mice into alymphoid RAG-2(o/o)/gammac(o/o) females. Recipients were mated and their implantation sites were examined by light microscopy, morphometry and ultrastructure. High numbers of uNK cells were established from each donor strain. Graft-derived uNK cells were similar in number and morphology to uNK cells of normal mice, suggesting that neither alpha- nor beta-ER is required for uNK precursor cell differentiation. Induction of spiral artery modification in the transplant recipients indicated that graft-derived uNK cells had functional properties. A novel technique for rapid isolation of highly purified uNK cells from normal mice using Dolichos biflorus agglutinin (DBA) lectin-conjugated magnetic beads was employed to obtain RNA. Expression of alpha- and beta-ER was absent by RT-PCR from NK cells isolated from the uterus, supporting the conclusions from the in vivo study.


Asunto(s)
Decidua/metabolismo , Células Asesinas Naturales/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Trasplante de Médula Ósea , Recuento de Células , Diferenciación Celular/fisiología , Células Cultivadas , Decidua/irrigación sanguínea , Decidua/citología , Implantación del Embrión/fisiología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Células Asesinas Naturales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Preñez , ARN Mensajero/metabolismo , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Mol Endocrinol ; 29(3): 281-6, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12459030

RESUMEN

The mouse knockout of the estrogen receptor alpha (ERalpha) gene, known as alphaERKO, has been extensively used for several years to study the role and function of ERalpha. Residual estradiol binding capacity in uterine tissue of 5-10% raised doubts if this knockout is a genuine null mutation of ERalpha. Although alternatively spliced ERalpha mRNA variants in the alphaERKO mouse were reported previously, the corresponding protein isoforms have not been detected to date. Here we show that a variant ERalpha protein, 61 kDa in size, is expressed in the uterine tissue of alphaERKO mice as a result of an alternative splicing. The transactivation capability of this protein is cell dependent and can be as high as 75% of the wild type ERalpha.


Asunto(s)
Eliminación de Gen , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Células 3T3 , Empalme Alternativo , Animales , Línea Celular , Receptor alfa de Estrógeno , Femenino , Células HeLa , Humanos , Ratones , Ratones Noqueados , Peso Molecular , Útero/metabolismo
16.
Proc Natl Acad Sci U S A ; 99(4): 2205-10, 2002 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-11854517

RESUMEN

Two isoforms of estrogen receptor (ER) have been described: ERalpha and ERbeta. The initial gene targeting of ERalpha, consisting in the introduction of a Neo cassette in exon 1 [alphaERKO, hereafter called ERalpha-Neo KO (knockout)], was reported in 1993. More recently, another mouse deficient in ERalpha because of the deletion of exon 2 (ERalphaKO, hereafter called ERalpha-delta2 KO) was generated. In ovariectomized ERalpha-wild-type mice, estradiol (E(2)) increases uterine weight and basal production of endothelial nitric oxide (NO). Both of these effects are abolished in ERalpha-delta2 KO mice. In contrast, we show here that both of these effects of E(2) are partially (uterine weight) or totally (endothelial NO production) preserved in ERalpha-Neo KO. We also confirm the presence of two ERalpha mRNA splice variants in uterus and aorta from ERalpha-Neo KO mice. One of them encodes a chimeric ERalpha protein (ERalpha55), partially deleted in the A/B domain, that was detected in both uterus and aorta by Western blot analysis. The other ERalpha mRNA splice variant codes for an isoform deleted for the A/B domain (ERalpha46), which was detected in uterus of ERalpha-Neo KO, and wild-type mice. This protein isoform was not detected in aorta. The identification of these two N-terminal modified isoforms in uterus, and at least one of them in aorta, probably explains the persistence of the E(2) effects in ERalpha-Neo KO mice. Furthermore, ERalpha-Neo KO mice may help in the elucidation of the specific functions of full-length ERalpha (ERalpha66) and ERalpha46, both shown to be physiologically generated in vivo.


Asunto(s)
Estradiol/farmacología , Óxido Nítrico/biosíntesis , Receptores de Estrógenos/genética , Receptores de Estrógenos/fisiología , Empalme Alternativo , Animales , Aorta/metabolismo , Western Blotting , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Receptor alfa de Estrógeno , Exones , Femenino , Hipertrofia , Inmunohistoquímica , Ratones , Ratones Noqueados , Modelos Genéticos , Mutagénesis Insercional , Tamaño de los Órganos , Isoformas de Proteínas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología
17.
Mol Endocrinol ; 15(12): 2064-77, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11731609

RESUMEN

The beneficial influence of E2 in the maintenance of healthy bone is well recognized. However, the way in which the actions of this hormone are mediated is less clearly understood. Western blot analysis of ERalpha in osteoblasts clearly demonstrated that the well characterized 66-kDa ERalpha was only one of the ERalpha isoforms present. Here we describe a 46-kDa isoform of ERalpha, expressed at a level similar to the 66-kDa isoform, that is also present in human primary osteoblasts. This shorter isoform is generated by alternative splicing of an ERalpha gene product, which results in exon 1 being skipped with a start codon in exon 2 used to initiate translation of the protein. Consequently, the transactivation domain AF-1 of this ERalpha isoform is absent. Functional analysis revealed that human (h)ERalpha46 is able to heterodimerize with the full-length ERalpha and also with ERbeta. Further, a DNA-binding complex that corresponds to hERalpha46 is detectable in human osteoblasts. We have shown that hERalpha46 is a strong inhibitor of hERalpha66 when they are coexpressed in the human osteosarcoma cell line SaOs. As a functional consequence, proliferation of the transfected cells is inhibited when increasing amounts of hERalpha46 are cotransfected with hERalpha66. In addition to human bone, the expression of the alternatively spliced ERalpha mRNA variant is also detectable in bone of ERalpha knockout mice. These data suggest that, in osteoblasts, E2 can act in part through an ERalpha isoform that is markedly different from the 66-kDa receptor. The expression of two ERalpha protein isoforms may account, in part, for the differential action that estrogens and estrogen analogs have in different tissues. In particular, the current models of the action of estrogens should be reevaluated to take account of the presence of at least two ERalpha protein isoforms in bone and perhaps in other tissues.


Asunto(s)
Osteoblastos/metabolismo , Receptores de Estrógenos/genética , Empalme Alternativo/genética , Southern Blotting , Electroforesis en Gel de Poliacrilamida , Receptor alfa de Estrógeno , Regulación de la Expresión Génica/genética , Humanos , Osteoblastos/fisiología , Pruebas de Precipitina , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/análisis , Receptores de Estrógenos/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Endonucleasas Específicas del ADN y ARN con un Solo Filamento/química , Transfección , Células Tumorales Cultivadas
18.
J Neurochem ; 79(4): 796-803, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11723172

RESUMEN

Cerebral apolipoprotein E (apoE) has been implicated in neuronal protection and repair. Due to the variable levels and types of estrogen receptors within different brain regions, the effect of estrogen on apoE and the mechanism of this effect may vary within different regions. Ovariectomized female C57BL/6 mice were treated with pharmacological levels of 17 beta-estradiol or placebo for 5 days, resulting in supraphysiological plasma levels of estradiol in the treated mice. ApoE and glial fibrillary acidic protein (GFAP) levels were measured in the cortex, hippocampus and diencephalon. 17 beta-Estradiol up-regulated apoE but not GFAP in the cortex and diencephalon, whereas in the hippocampus, GFAP and apoE were equally up-regulated. Treatment of estrogen receptor (ER) alpha knockout mice with 17 beta-estradiol or treatment of C57BL/6 mice with 17 alpha-estradiol, a poor estrogen receptor agonist, specifically induced apoE in the cortex, but not in the diencephalon. These results indicate that 17 beta-estradiol effects on apoE are either directly or indirectly mediated by ER alpha in the diencephalon, while the effects in the cortex may be mediated by a non-classical mechanism or by ER beta. Measurement of mRNA levels in estrogen versus placebo-treated wild-type mice indicated that the effect of 17 beta-estradiol on apoE was not associated with changes in apoE mRNA levels.


Asunto(s)
Apolipoproteínas E/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Estradiol/farmacología , Animales , Apolipoproteínas E/genética , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Diencéfalo/efectos de los fármacos , Diencéfalo/metabolismo , Implantes de Medicamentos , Estradiol/sangre , Receptor alfa de Estrógeno , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos/efectos de los fármacos , Ovariectomía , ARN Mensajero/metabolismo , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Regulación hacia Arriba/efectos de los fármacos
19.
Endocrinology ; 142(12): 5145-8, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11713208

RESUMEN

In neocortical explants, E2 activates various signaling components of the MAPK cascade, including B-Raf and MAPK kinase-dependent ERK, suggesting a possible role in the differentiative actions of E2 in the brain. To further characterize the signaling pathways activated by E2, we determined whether c-Src, a member of the Src family of nonreceptor tyrosine kinases and an important modulator of both the MAPK cascade and neuronal differentiation, may play a role in E2 signaling. The present studies show for the first time in the brain that E2 elicits phosphorylation of c-Src on three functionally critical tyrosine residues (Y220, Y423, and Y534), and that this phosphorylation occurs despite disruption of ER alpha (in ER knockout mice). PP2, a Src family kinase inhibitor, suppressed not only E2-induced phosphorylation of c-Src, but ERK phosphorylation as well, suggesting that c-Src may be an upstream regulator of E2 signaling. E2-induced phosphorylation of c-Src is associated with increased tyrosine phosphorylation of Shc, increased association of Shc with Grb2, and induction of Ras, but not Rap1, activation. Together, these data provide evidence that E2 activates a novel c-Src-dependent signal transduction pathway in the developing brain.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Estradiol/farmacología , Neocórtex/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Proteínas de Unión al ADN/farmacología , Activación Enzimática , Proteína Adaptadora GRB2 , Genes src , Transferasas Intramoleculares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Proteínas de Plantas/farmacología , Proteínas/metabolismo , Valores de Referencia , Proteínas de Unión al GTP rap1/fisiología , Proteínas ras/fisiología , Familia-src Quinasas
20.
J Appl Physiol (1985) ; 91(5): 2391-9; discussion 2389-90, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11641386

RESUMEN

Estrogen alters reactivity of cerebral arteries by modifying production of endothelium-dependent vasodilators. Estrogen receptors (ER) are thought to be involved, but the responsible ER subtype is unknown. ER-alpha knockout (alphaERKO) mice were used to test whether estrogen acts via ER-alpha. Mice were ovariectomized, with or without estrogen replacement, and cerebral blood vessels were isolated 1 mo later. Estrogen increased levels of endothelial nitric oxide synthase and cyclooxygenase-1 in vessels from wild-type mice but was ineffective in alphaERKO mice. Endothelium-denuded middle cerebral artery segments from all animals constricted when pressurized. In denuded arteries from alphaERKO but not wild-type mice, estrogen treatment enhanced constriction. In endothelium-intact, pressurized arteries from wild-type estrogen-treated mice, diameters were larger compared with arteries from untreated wild-type mice. In addition, contractile responses to indomethacin were greater in arteries from wild-type estrogen-treated mice compared with arteries from untreated wild-type mice. In contrast, estrogen treatment of alphaERKO mice had no effect on diameter or indomethacin responses of endothelium-intact arteries. Thus ER-alpha regulation of endothelial nitric oxide synthase and cyclooxygenase-1 pathways appears to contribute to effects of estrogen on cerebral artery reactivity.


Asunto(s)
Óxido Nítrico Sintasa/biosíntesis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Receptores de Estrógenos/genética , Animales , Western Blotting , Peso Corporal/fisiología , Arterias Cerebrales/enzimología , Inhibidores de la Ciclooxigenasa/farmacología , Electroforesis en Gel de Poliacrilamida , Endotelio Vascular/enzimología , Receptor alfa de Estrógeno , Femenino , Masculino , Ratones , Músculo Liso Vascular/fisiología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Tamaño de los Órganos/fisiología , Ovariectomía , Prostaglandina-Endoperóxido Sintasas/genética , Caracteres Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA