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

Banco 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.
Nat Med ; 3(5): 545-8, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9142124

RESUMEN

The atheroprotective effects of estrogen in women are well recognized, but the underlying mechanisms responsible are not well understood. Blood vessel cells express the classic estrogen receptor, ER alpha (ref. 2-6), and are directly affected by estrogen, which inhibits the development of atherosclerotic and injury-induced vascular lesions. We have generated mice in which the ER alpha gene is disrupted and have used a mouse model of carotid arterial injury to compare the effects of estrogen on wild-type and estrogen receptor-deficient mice. Increases in vascular medial area and smooth muscle cell proliferation were quantified following vascular injury in ovariectomized mice treated with vehicle or with physiologic levels of 17 beta-estradiol. Surprisingly, in both wild-type and estrogen receptor-deficient mice, 17 beta-estradiol markedly inhibited to the same degree all measures of vascular injury. These data demonstrate that estrogen inhibits vascular by a novel mechanism that is independent of the classic estrogen receptor, ER alpha.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Estradiol/farmacología , Músculo Liso Vascular/efectos de los fármacos , Receptores de Estrógenos/fisiología , Animales , Arterias Carótidas , División Celular , Endotelio Vascular/patología , Femenino , Expresión Génica , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Receptores de Estrógenos/genética
3.
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
4.
Science ; 266(5190): 1524-7, 1994 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-7985022

RESUMEN

Estrogen hormones produce physiological actions within a variety of target sites in the body and during development by activating a specific receptor protein. Hormone responsiveness for the estrogen receptor protein was investigated at different stages of development with the use of gene knockout techniques because no natural genetic mutants have been described. A mutant mouse line without a functional estrogen receptor was created and is being used to assess estrogen responsiveness. Both sexes of these mutant animals are infertile and show a variety of phenotypic changes, some of which are associated with the gonads, mammary glands, reproductive tracts, and skeletal tissues.


Asunto(s)
Estrógenos/fisiología , Receptores de Estrógenos/fisiología , Animales , Femenino , Heterocigoto , Homocigoto , Humanos , Infertilidad Femenina/etiología , Infertilidad Masculina/etiología , Masculino , Ratones , Ratones Noqueados , Mutación , Fenotipo , Receptores de Estrógenos/genética , Transducción de Señal
5.
Science ; 286(5448): 2328-31, 1999 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-10600740

RESUMEN

Mice lacking estrogen receptors alpha and beta were generated to clarify the roles of each receptor in the physiology of estrogen target tissues. Both sexes of alphabeta estrogen receptor knockout (alphabetaERKO) mutants exhibit normal reproductive tract development but are infertile. Ovaries of adult alphabetaERKO females exhibit follicle transdifferentiation to structures resembling seminiferous tubules of the testis, including Sertoli-like cells and expression of Müllerian inhibiting substance, sulfated glycoprotein-2, and Sox9. Therefore, loss of both receptors leads to an ovarian phenotype that is distinct from that of the individual ERKO mutants, which indicates that both receptors are required for the maintenance of germ and somatic cells in the postnatal ovary.


Asunto(s)
Trastornos del Desarrollo Sexual , Chaperonas Moleculares , Ovario/anatomía & histología , Ovario/fisiología , Receptores de Estrógenos/fisiología , Animales , Hormona Antimülleriana , Diferenciación Celular , Clusterina , Estradiol/fisiología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Marcación de Gen , Glicoproteínas/análisis , Inhibidores de Crecimiento/análisis , Proteínas del Grupo de Alta Movilidad/análisis , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Noqueados , Ovario/citología , Ovario/crecimiento & desarrollo , Receptores de Estrógenos/genética , Factor de Transcripción SOX9 , Túbulos Seminíferos/anatomía & histología , Túbulos Seminíferos/citología , Células de Sertoli/citología , Transducción de Señal , Hormonas Testiculares/análisis , Testículo/anatomía & histología , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/fisiología , Factores de Transcripción/análisis
6.
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
7.
Endocr Rev ; 20(3): 358-417, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10368776

RESUMEN

All scientific investigations begin with distinct objectives: first is the hypothesis upon which studies are undertaken to disprove, and second is the overall aim of obtaining further information, from which future and more precise hypotheses may be drawn. Studies focusing on the generation and use of gene-targeted animal models also apply these goals and may be loosely categorized into sequential phases that become apparent as the use of the model progresses. Initial studies of knockout models often focus on the plausibility of the model based on prior knowledge and whether the generation of an animal lacking the particular gene will prove lethal or not. Upon the successful generation of a knockout, confirmatory studies are undertaken to corroborate previously established hypotheses of the function of the disrupted gene product. As these studies continue, observations of unpredicted phenotypes or, more likely, the lack of a phenotype that was expected based on models put forth from past investigations are noted. Often the surprising phenotype is due to the loss of a gene product that is downstream from the functions of the disrupted gene, whereas the lack of an expected phenotype may be due to compensatory roles filled by alternate mechanisms. As the descriptive studies of the knockout continue, use of the model is often shifted to the role as a unique research reagent, to be used in studies that 1) were not previously possible in a wild-type model; 2) aimed at finding related proteins or pathways whose existence or functions were previously masked; or 3) the subsequent effects of the gene disruption on related physiological and biochemical systems. The alpha ERKO mice continue to satisfy the confirmatory role of a knockout quite well. As summarized in Table 4, the phenotypes observed in the alpha ERKO due to estrogen insensitivity have definitively illustrated several roles that were previously believed to be dependent on functional ER alpha, including 1) the proliferative and differentiative actions critical to the function of the adult female reproductive tract and mammary gland; 2) as an obligatory component in growth factor signaling in the uterus and mammary gland; 3) as the principal steroid involved in negative regulation of gonadotropin gene transcription and LH levels in the hypothalamic-pituitary axis; 4) as a positive regulator of PR expression in several tissues; 5) in the positive regulation of PRL synthesis and secretion from the pituitary; 6) as a promotional factor in oncogene-induced mammary neoplasia; and 7) as a crucial component in the differentiation and activation of several behaviors in both the female and male. The list of unpredictable phenotypes in the alpha ERKO must begin with the observation that generation of an animal lacking a functional ER alpha gene was successful and produced animals of both sexes that exhibit a life span comparable to wild-type. The successful generation of beta ERKO mice suggests that this receptor is also not essential to survival and was most likely not a compensatory factor in the survival of the alpha ERKO. In support of this is our recent successful generation of double knockout, or alpha beta ERKO mice of both sexes. The precise defects in certain components of male reproduction, including the production of abnormal sperm and the loss of intromission and ejaculatory responses that were observed in the alpha ERKO, were quite surprising. In turn, certain estrogen pathways in the alpha ERKO female appear intact or unaffected, such as the ability of the uterus to successfully exhibit a progesterone-induced decidualization response, and the possible maintenance of an LH surge system in the hypothalamus. [ABSTRACT TRUNCATED]


Asunto(s)
Ratones Noqueados , Receptores de Estrógenos/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Noqueados/genética , Ratones Noqueados/fisiología , Sistemas Neurosecretores/fisiología , Fenotipo , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Sistema Urogenital/fisiología
8.
J Clin Invest ; 107(3): 333-40, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11160157

RESUMEN

The inhibitory effects of estrogen (17beta-estradiol) on atherosclerosis have been well documented in numerous animal models, and epidemiological evidence supports this protective effect in humans. The detailed mechanisms for this protection are not understood, but most are thought to be mediated through estrogen receptors (ERs), of which two are known (ERalpha and ERbeta). To investigate the role of ERalpha in the atheroprotective effect of 17beta-estradiol (E2), we ovariectomized female mice that lack apoE (AAee) or lack both apoE and ERalpha (alphaalphaee), and treated half of them with E2 for three months. E2 treatment of ovariectomized AAee females dramatically reduced the size of the lesions as well as their histological complexity. Plasma cholesterol was significantly reduced in this group, although the observed extent of protection by E2 was greater than could be explained solely by the change in lipid levels. In contrast, E2 treatment of ovariectomized alphaalphaee females caused minimal reduction in lesion size and no reduction in total plasma cholesterol compared with alphaalphaee mice without E2, demonstrating that ERalpha is a major mediator of the atheroprotective effect of E2. Nevertheless, E2 treatment significantly reduced the complexity of plaques in the alphaalphaee females, although not to the same degree as in AAee females, suggesting the existence of ERalpha-independent atheroprotective effects of E2.


Asunto(s)
Apolipoproteínas E/genética , Estradiol/farmacología , Receptores de Estrógenos/fisiología , Animales , Apolipoproteínas E/deficiencia , Arteriosclerosis/sangre , Arteriosclerosis/patología , Arteriosclerosis/prevención & control , Peso Corporal , Colorantes , Preparaciones de Acción Retardada , Estradiol/administración & dosificación , Estradiol/sangre , Receptor alfa de Estrógeno , Femenino , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Ovariectomía , Seno Aórtico , Piel/patología , Útero/patología
9.
J Clin Invest ; 99(10): 2429-37, 1997 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9153286

RESUMEN

The present study was designed to test the hypothesis that estrogen receptors (ER) in the blood vessel wall play a role in the modulation of the release of endothelium-derived nitric oxide (EDNO). Both basal and stimulated release of EDNO were determined in aortic rings isolated from female and male wild-type and male homozygous estrogen receptor knock-out (ERKO) mice. 125I-17beta-estradiol binding in aortic tissue showed significantly more high affinity cytosolic- nuclear-binding sites in male compared with female wildtype mice. Estrogen receptor transcripts were present in the aorta of male wild-type mice, but they were absent in male ERKO animals. Basal release of EDNO (determined by endothelium-dependent contraction caused by NG-nitro-arginine) was significantly higher in aorta of wild-type male mice compared with wild-type female mice, and significantly lower in the aorta of male ERKO compared with male wild-type mice. Acetylcholine-induced endothelium-dependent relaxation was similar in all groups studied. No difference was observed in the activity of calcium-dependent nitric oxide synthase in homogenates of lungs and brain taken from male wild-type and ERKO mice. These studies show a significant association between the number of estrogen receptors and basal release of EDNO in the aorta of mice, and suggest that decreased vascular estrogen receptor number may represent a novel risk factor for cardiovascular diseases.


Asunto(s)
Endotelio Vascular/fisiología , Músculo Liso Vascular/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/biosíntesis , Nitroarginina/farmacología , Receptores de Estrógenos/biosíntesis , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animales , Aorta/fisiología , Encéfalo/enzimología , Cartilla de ADN , Endotelio Vascular/efectos de los fármacos , Inducción Enzimática , Estradiol/sangre , Exones , Femenino , Técnicas In Vitro , Pulmón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Endoperóxidos de Prostaglandinas Sintéticos/farmacología , Receptores de Estrógenos/genética , Caracteres Sexuales , Especificidad de la Especie , Tromboxano A2/análogos & derivados , Tromboxano A2/farmacología , Vasoconstrictores/farmacología
10.
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
11.
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
12.
Cancer Res ; 60(24): 6901-10, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11156389

RESUMEN

Although several lines of epidemiological evidence suggest that estrogen exposure influences the incidence of breast cancer development, the mechanisms by which estrogen may stimulate the formation of breast cancer remain poorly understood. We have explored how alterations in estrogen exposure can influence the development of mammary cancer in the C3(1)/T(AG) transgenic model, where estrogen levels and estrogen receptor alpha (ERalpha) expression do not appear to modify the level of transgene expression. The C3(1)/T(AG) transgene becomes transcriptionally active in mammary ductal target cells at 3 weeks of age after the estrogen-induced differentiation of the mammary epithelial anlage to the ductal outgrowth stage. Complete maturation of the mammary ductal tree, however, is not required for cancer development because tumors arise in animals where ductal branching and terminal end bud formation have been prematurely arrested by ovariectomy. Mammary tumorigenesis in this model is promoted by increased estrogen exposure with the development of significantly more mammary intraepithelial neoplastic lesions and carcinomas associated with accelerated malignant conversion. The promotion of mammary tumors in this model appears to occur through an estrogen-induced proliferation and increase in the number of available target cells for transformation at the terminal ductal lobular units, as has been postulated to occur in women who receive hormone replacement therapy and/or by additional molecular mechanisms. We show, for the first time in a transgenic mouse model, that mammary tumor progression is associated with the loss of ERalpha expression, as has been often observed in human breast cancers with important clinical significance. Estrogen signaling may, therefore, serve different functions, depending upon the stage of tumorigenesis. ERbeta expression is up-regulated during tumor progression, although the functional significance of this remains to be determined.


Asunto(s)
Antígenos Virales de Tumores/genética , Estrógenos/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones Transgénicos , Receptores de Estrógenos/biosíntesis , Animales , Northern Blotting , Western Blotting , Progresión de la Enfermedad , Receptor alfa de Estrógeno , Estrógenos/farmacología , Femenino , Inmunohistoquímica , Neoplasias Mamarias Experimentales/genética , Ratones , Mutación , Fosforilación , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Distribución Tisular , Transfección , Células Tumorales Cultivadas
13.
Cancer Res ; 59(8): 1869-76, 1999 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10213494

RESUMEN

Estrogens have important functions in mammary gland development and carcinogenesis. To better define these roles, we have used two previously characterized lines of genetically altered mice: estrogen receptor-alpha (ER alpha) knockout (ERKO) mice, which lack the gene encoding ER alpha, and mouse mammary virus tumor (MMTV)-Wnt-1 transgenic mice (Wnt-1 TG), which develop mammary hyperplasia and neoplasia due to ectopic production of the Wnt-1 secretory glycoprotein. We have crossed these lines to ascertain the effects of ER alpha deficiency on mammary gland development and carcinogenesis in mice expressing the Wnt-1 transgene. Introduction of the Wnt-1 transgene into the ERKO background stimulates proliferation of alveolar-like epithelium, indicating that Wnt-1 protein can promote mitogenesis in the absence of an ER alpha-mediated response. The hyperplastic glandular tissue remains confined to the nipple region, implying that the requirement for ER alpha in ductal expansion is not overcome by ectopic Wnt-1. Tumors were detected in virgin ERKO females expressing the Wnt-1 transgene at an average age (48 weeks) that is twice that seen in virgin Wnt-1 TG mice (24 weeks) competent to produce ER alpha. Prepubertal ovariectomy of Wnt-1 TG mice also extended tumor latency to 42 weeks. However, pregnancy did not appear to accelerate the appearance of tumors in Wnt-1 TG mice, and tumor growth rates were not measurably affected by late ovariectomy. Small hyperplastic mammary glands were observed in Wnt-1 TG males, regardless of ER alpha gene status; the glands were similar in appearance to those found in ERKO/Wnt-1 TG females. Mammary tumors also occurred in Wnt-1 TG males; latency tended to be longer in the heterozygous ER alpha and ERKO males (86 to 100 weeks) than in wild-type ER alpha mice (ca. 75 weeks). We conclude that ectopic expression of the Wnt-1 proto-oncogene can induce mammary hyperplasia and tumorigenesis in the absence of ER alpha in female and male mice. The delayed time of tumor appearance may depend on the number of cells at risk of secondary events in the hyperplastic glands, on the carcinogenesis-promoting effects of ER alpha signaling, or on both.


Asunto(s)
Mama/patología , Neoplasias Mamarias Animales/genética , Virus del Tumor Mamario del Ratón/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Pez Cebra , Animales , Transformación Celular Viral , Receptor alfa de Estrógeno , Estrógenos/metabolismo , Femenino , Eliminación de Gen , Técnicas de Transferencia de Gen , Hiperplasia , Masculino , Neoplasias Mamarias Animales/patología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/biosíntesis , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Transformación Genética , Proteínas Wnt , Proteína Wnt1
14.
Cancer Res ; 61(16): 6089-97, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11507058

RESUMEN

Neonatal exposure of rodents to high doses of estrogen permanently imprints the growth and function of the prostate and predisposes this gland to hyperplasia and severe dysplasia analogous to prostatic intraepithelial neoplasia with aging. Because the rodent prostate gland expresses estrogen receptor (ER)-alpha within a subpopulation of stromal cells and ERbeta within epithelial cells, the present study was undertaken to determine the specific ER(s) involved in mediating prostatic developmental estrogenization. Wild-type (WT) mice, homozygous mutant ER (ERKO) alpha -/- mice, and betaERKO -/- mice were injected with 2 microg of diethylstilbestrol (DES) or oil (controls) on days 1, 3, and 5 of life. Reproductive tracts were excised on days 5 or 10 (prepubertal), day 30 (pubertal), day 90 (young adult), or with aging at 6, 12, and 18 months of age. Prostate complexes were microdissected and examined histologically for prostatic lesions and markers of estrogenization. Immunocytochemistry was used to examine expression of androgen receptor, ERalpha, ERbeta, cytokeratin 14 (basal cells), cytokeratin 18 (luminal cells), and dorsolateral protein over time in the treated mice. In WT-DES mice, developmental estrogenization of the prostate was observed at all of the time points as compared with WT-oil mice. These prostatic imprints included transient up-regulation of ERalpha, down-regulation of androgen receptor, decreased ERbeta levels in adult prostate epithelium, lack of DLP secretory protein, and a continuous layer of basal cells lining the ducts. With aging, epithelial dysplasia and inflammatory cell infiltrate were observed in the ventral and dorsolateral prostate lobes. In contrast, the prostates of alphaERKO mice exhibited no response to neonatal DES either immediately after exposure or throughout life up to 18 months of age. Furthermore, neonatal DES treatment of betaERKO mice resulted in a prostatic response similar to that observed in WT animals. The present findings indicate that ERalpha is the dominant ER form mediating the developmental estrogenization of the prostate gland. If epithelial ERbeta is involved in some component of estrogen imprinting, its role would be considered minor and would require the presence of ERalpha expression in the prostatic stromal cells.


Asunto(s)
Dietilestilbestrol/farmacología , Estrógenos/fisiología , Próstata/efectos de los fármacos , Próstata/crecimiento & desarrollo , Receptores de Estrógenos/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
15.
Biochim Biophys Acta ; 992(1): 1-8, 1989 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-2752033

RESUMEN

Heat shock protein 90 was purified from calf uterus and rat liver. Both heat shock protein 90s had similar molecular weights, as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, of Mr 87,000 and 88,000, isoelectric points of 5.2, and Stokes radii of 6.7 and 6.5 nm, respectively. Heat shock protein 90 bound to phenyl-Sepharose CL-4B even at low ionic strength, and also bound to butyl-Toyopearl at high ionic strength. Heat shock protein 90 bound to phenyl-Sepharose could be eluted with a buffer containing organic solvents or detergents such as 2-propanol, dioxane, dimethylformamide, methyl cellosolve, 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate or Triton X-100, but not with ionic salts such as 1 M KCl. These results suggest that heat shock protein 90 possesses a significant hydrophobic region on the surface of the molecule. Hydrophobicities of heat shock protein 90 and 4S calf uterine estrogen receptor were both decreased by formation of a 8 S estrogen receptor complex. The role of the hydrophobic region of heat shock protein 90 in the interaction with estrogen receptor and other proteins is discussed.


Asunto(s)
Proteínas de Choque Térmico/aislamiento & purificación , Hígado/metabolismo , Útero/metabolismo , Animales , Bovinos , Cromatografía en Gel , Electroforesis en Gel Bidimensional , Femenino , Conformación Proteica , Ratas , Ratas Endogámicas , Receptores de Estrógenos/análisis
16.
J Gen Physiol ; 110(2): 135-40, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9236206

RESUMEN

Steroid hormones control the expression of many cellular regulators, and a role for estrogen in cardiovascular function and disease has been well documented. To address whether the activity of the L-type Ca2+ channel, a critical element in cardiac excitability and contractility, is altered by estrogen and its nuclear receptor, we examined cardiac myocytes from male mice in which the estrogen receptor gene had been disrupted (ERKO mice). Binding of dihydropyridine Ca2+ channel antagonist isradipine (PN200-110) was increased 45.6% in cardiac membranes from the ERKO mice compared to controls, suggesting that a lack of estrogen receptors in the heart increased the number of Ca2+ channels. Whole-cell patch clamp of acutely dissociated adult cardiac ventricular myocytes indicated that Ca2+ channel current was increased by 49% and action potential duration was increased by 75%. Examination of electrocardiogram parameters in ERKO mice showed a 70% increase in the QT interval without significant changes in PQ or QRS intervals. These results show that the membrane density of the cardiac L-type Ca2+ channel is regulated by the estrogen receptor and suggest that decreased estrogen may lead to an increase in the number of cardiac L-type Ca2+ channels, abnormalities in cardiac excitability, and increased risk of arrhythmia and cardiovascular disease.


Asunto(s)
Canales de Calcio/metabolismo , Miocardio/metabolismo , Receptores de Estrógenos/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/metabolismo , Estimulación Eléctrica , Electrofisiología , Isradipino/farmacología , Ligandos , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Técnicas de Placa-Clamp , Receptores de Estrógenos/fisiología , Factores de Transcripción/fisiología
17.
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
18.
Trends Endocrinol Metab ; 8(4): 137-45, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-18406799

RESUMEN

Disruption of the estrogen receptor (ER) gene in mice causes infertility in both sexes. Infertility in female ER knockout (ERKO) mice results from altered development of accessory sex structures, disrupted endocrine physiology, and disrupted gametogenesis. Male accessory sex structures appear relatively normal, with infertility stemming from altered sexual behaviors and disrupted gametogenesis. These findings provide significant insights into the biological importance of the ER and suggest further areas for examining the impact of estrogens on reproductive biology.

19.
J Mol Med (Berl) ; 76(7): 497-511, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9660168

RESUMEN

Decades of study have described a number roles fulfilled by the steroid hormones and their respective receptors in sexual differentiation and development, reproductive function and behavior, and more recently in the function and maintenance of non-reproductive organ systems, such as skeletal muscle, bone and coronary tissues. The biological effects of the steroid hormones are believed to be mediated in part by specific receptor proteins that demonstrated great specificity for their respective steroid ligands. Much of the experimental research of the functions of the sex steroid receptors has depended upon in vitro systems as well as in vivo methods that require surgical castration or the pharmacological administration of hormone antagonists. However, recently developed techniques that allow for manipulation of the mouse genome have been utilized to generate transgenic animals that lack functional estrogen or progesterone receptors. These transgenic animals, combined with the naturally existing Tfm mice which lack functional androgen receptor, now provide in vivo models for further study of the various actions of the sex steroids and their receptors. This review attempts to describe and compare the various phenotypes that result in each of these lines of mice, with emphasis on the development and function of the reproductive systems as well as reproductive behavior.


Asunto(s)
Receptores de Esteroides/fisiología , Transducción de Señal/fisiología , Esteroides/fisiología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos
20.
Curr Opin Pharmacol ; 1(6): 613-9, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11757817

RESUMEN

The estrogen receptors ERalpha and ERbeta are the main mediators of estrogen action and estrogens play an important role in a variety of aspects of physiology besides their well acknowledged function in reproduction. In vivo and in vitro studies indicate that the estrogen receptors are mechanistically implicated in endocrine-related diseases. Recent studies with estrogen receptor knockout mice have helped to unravel the role of the estrogen receptors in brain degeneration, osteoporosis, cardiovascular diseases and obesity.


Asunto(s)
Enfermedades del Sistema Endocrino/etiología , Receptores de Estrógenos/fisiología , Tejido Adiposo/metabolismo , Animales , Huesos/metabolismo , Encéfalo/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Enfermedades del Sistema Endocrino/metabolismo , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Osteoporosis/etiología , Osteoporosis/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA