Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(2): E283-90, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24371309

RESUMEN

Estrogen receptor alpha (ERα) activation functions AF-1 and AF-2 classically mediate gene transcription in response to estradiol (E2). A fraction of ERα is targeted to plasma membrane and elicits membrane-initiated steroid signaling (MISS), but the physiological roles of MISS in vivo are poorly understood. We therefore generated a mouse with a point mutation of the palmitoylation site of ERα (C451A-ERα) to obtain membrane-specific loss of function of ERα. The abrogation of membrane localization of ERα in vivo was confirmed in primary hepatocytes, and it resulted in female infertility with abnormal ovaries lacking corpora lutea and increase in luteinizing hormone levels. In contrast, E2 action in the uterus was preserved in C451A-ERα mice and endometrial epithelial proliferation was similar to wild type. However, E2 vascular actions such as rapid dilatation, acceleration of endothelial repair, and endothelial NO synthase phosphorylation were abrogated in C451A-ERα mice. A complementary mutant mouse lacking the transactivation function AF-2 of ERα (ERα-AF2(0)) provided selective loss of function of nuclear ERα actions. In ERα-AF2(0), the acceleration of endothelial repair in response to estrogen-dendrimer conjugate, which is a membrane-selective ER ligand, was unaltered, demonstrating integrity of MISS actions. In genome-wide analysis of uterine gene expression, the vast majority of E2-dependent gene regulation was abrogated in ERα-AF2(0), whereas in C451A-ERα it was nearly fully preserved, indicating that membrane-to-nuclear receptor cross-talk in vivo is modest in the uterus. Thus, this work genetically segregated membrane versus nuclear actions of a steroid hormone receptor and demonstrated their in vivo tissue-specific roles.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrógeno/genética , Ovario/fisiología , Útero/metabolismo , Análisis de Varianza , Animales , Western Blotting , Movimiento Celular , Biología Computacional , Células Endoteliales , Receptor alfa de Estrógeno/metabolismo , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Lipoilación/genética , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Ovario/metabolismo , Mutación Puntual/genética , Receptor Cross-Talk/fisiología
2.
Am J Physiol Endocrinol Metab ; 310(11): E912-8, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27048997

RESUMEN

The bone-sparing effect of estrogens is mediated primarily via estrogen receptor (ER)α, which stimulates gene transcription through activation function (AF)-1 and AF-2. The role of ERαAF-1 for the estradiol (E2) effects is tissue specific. The selective ER modulators (SERMs) raloxifene (Ral), lasofoxifene (Las), and bazedoxifene (Bza) can be used to treat postmenopausal osteoporosis. They all reduce the risk for vertebral fractures, whereas Las and partly Bza, but not Ral, reduce the risk for nonvertebral fractures. Here, we have compared the tissue specificity of Ral, Las, and Bza and evaluated the role of ERαAF-1 for the effects of these SERMs, with an emphasis on bone parameters. We treated ovariectomized (OVX) wild-type (WT) mice and OVX mice lacking ERαAF-1 (ERαAF-1(0)) with E2, Ral, Las, or Bza. All three SERMs increased trabecular bone mass in the axial skeleton. In the appendicular skeleton, only Las increased the trabecular bone volume/tissue volume and trabecular number, whereas both Ral and Las increased the cortical bone thickness and strength. However, Ral also increased cortical porosity. The three SERMs had only a minor effect on uterine weight. Notably, all evaluated effects of these SERMs were absent in ovx ERαAF-1(0) mice. In conclusion, all SERMs had similar effects on axial bone mass. However, the SERMs had slightly different effects on the appendicular skeleton since only Las increased the trabecular bone mass and only Ral increased the cortical porosity. Importantly, all SERM effects require a functional ERαAF-1 in female mice. These results could lead to development of more specific treatments for osteoporosis.


Asunto(s)
Densidad Ósea/fisiología , Moduladores de los Receptores de Estrógeno/administración & dosificación , Receptor alfa de Estrógeno/metabolismo , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/fisiología , Animales , Densidad Ósea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/fisiología , Ovariectomía , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
3.
J Immunol ; 190(11): 5459-70, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23626011

RESUMEN

17ß-Estradiol (E2) has been shown to regulate GM-CSF- or Flt3 ligand-driven dendritic cell (DC) development through estrogen receptor (ER) α signaling in myeloid progenitors. ERα regulates transcription of target genes through two distinct activation functions (AFs), AF-1 and AF-2, whose respective involvement varies in a cell type- or tissue-specific manner. In this study, we investigated the role of ERα AFs in the development and effector functions of inflammatory DCs, steady-state conventional DCs, and plasmacytoid DCs (pDC), using mouse lacking either AF-1 or AF-2. In agreement with previous works, we showed that E2 fostered the differentiation and effector functions of inflammatory DCs through ERα-dependent upregulation of IFN regulatory factor (IRF)-4 in GM-CSF-stimulated myeloid progenitors. Interestingly, whereas AF-1 was required for early IRF-4 upregulation in DC precursors, it was dispensable to enhance IRF-4 expression in differentiated DCs to a level compatible with the development of the more functional Ly6C(-) CD11b(+) DC subset. Presence of E2 had no effect on progenitors from either knock-in mice with 7-aa deletion in helix 12 of ERα, lacking AF-2, or ERα(-/-) mice. By contrast, in Flt3 ligand-driven DC differentiation, activation of AF-1 domain was required to promote the development of more functionally competent conventional DCs and pDCs. Moreover, lack of ERα AF-1 blunted the TLR7-mediated IFN-α response of female pDCs in vivo. Thus, our study demonstrates that ERα uses AF-1 differently in steady-state and inflammatory DC lineages to regulate their innate functions, suggesting that selective ER modulators could be used to target specific DC subsets.


Asunto(s)
Células Dendríticas/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Dominios y Motivos de Interacción de Proteínas , Animales , Antígenos CD11/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Receptor alfa de Estrógeno/química , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Transgénicos , Fenotipo , Transducción de Señal , Receptores Toll-Like/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(48): 19822-7, 2012 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-23150547

RESUMEN

We previously generated and characterized a genuine estrogen receptor (ER) ß-null mouse line (named ERß(ST)(L-/L-)) and showed that ERß(ST)(L-/L-) mice were sterile, due to an ovulation impairment in females and to an unknown reason in males, as their reproductive organs and spermatozoid motility appeared normal. We report here an assessment of the sexual behavior of ERß(ST)(L-/L-) null mice. We found that ERß(ST)(L-/L-) males display mildly impaired sexual behavior and that ERß(ST)(L-/L-) females are significantly less receptive and less attractive than wild-type (WT) females. Decreased attractivity is also exhibited by ERßAF2(0) but not by ERßAF1(0) mutant females (females devoid of either AF2 or AF1 activation function of ERß). Interestingly, by using an odor preference test, we have determined that the low attractiveness of ERß(ST)(L-/L-) and ERßAF2(0) females is related to a deficiency of a volatile chemosignal.


Asunto(s)
Receptor beta de Estrógeno/fisiología , Estrógenos/fisiología , Conducta Sexual Animal , Animales , Receptor beta de Estrógeno/genética , Femenino , Masculino , Ratones , Ratones Noqueados
5.
Proc Natl Acad Sci U S A ; 109(3): 983-8, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22215598

RESUMEN

It has generally been assumed that bone mass is controlled by endocrine mechanisms and the local bone environment. Recent findings demonstrate that central pathways are involved in the regulation of bone mass. Estrogen is involved in the regulation of bone homeostasis and the CNS is also a target for estrogen actions. The aim of this study was to investigate in vivo the role of central estrogen receptor-α (ERα) expression for bone mass. Nestin-Cre mice were crossed with ERα(flox) mice to generate mice lacking ERα expression specifically in nervous tissue (nestin-ERα(-/-)). Bone mineral density was increased in both the trabecular and cortical bone compartments in nestin-ERα(-/-) mice compared with controls. Femoral bone strength was increased in nestin-ERα(-/-) mice, as demonstrated by increased stiffness and maximal load of failure. The high bone mass phenotype in nestin-ERα(-/-) mice was mainly caused by increased bone formation. Serum leptin levels were elevated as a result of increased leptin expression in white adipose tissue (WAT) and slightly increased amount of WAT in nestin-ERα(-/-) mice. Leptin receptor mRNA levels were reduced in the hypothalamus but not in bone. In conclusion, inactivation of central ERα signaling results in increased bone mass, demonstrating that the balance between peripheral stimulatory and central inhibitory ERα actions is important for the regulation of bone mass. We propose that the increased bone mass in nestin-ERα(-/-) mice is mediated via decreased central leptin sensitivity and thereby increased secretion of leptin from WAT, which, in turn, results in increased peripheral leptin-induced bone formation.


Asunto(s)
Huesos/metabolismo , Huesos/patología , Receptor alfa de Estrógeno/metabolismo , Neuronas/metabolismo , Animales , Densidad Ósea , Remodelación Ósea , Huesos/diagnóstico por imagen , Huesos/cirugía , Receptor alfa de Estrógeno/deficiencia , Femenino , Hormona Folículo Estimulante/metabolismo , Eliminación de Gen , Proteínas de Filamentos Intermediarios/metabolismo , Leptina/sangre , Hormona Luteinizante/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Nestina , Tamaño de los Órganos , Ovariectomía , Radiografía , Serotonina/metabolismo , Transducción de Señal , Esteroides/metabolismo , Linfocitos T/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(32): 13311-6, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788522

RESUMEN

17ß-Estradiol (E2) regulates estrogen receptor-α (ERα) target gene transcription through the two independent activation functions (AFs), AF1 and AF2, located in the N-terminal and ligand binding domain of ERα, respectively. We previously reported that ERα is required for the E2 atheroprotective action as well as for its accelerative action on endothelial healing, but its AF1 function is dispensable. Here, we investigated the role of ERαAF2 in these two major beneficial actions of E2 by electively targeting ERαAF2 (named ERαAF2(0)). Our results prove four points. (i) Compared with WT ERα, the ability of ERαAF2(0) to stimulate the C3 complement or the estrogen response element-thymidine kinase promoter in two cell lines was dramatically decreased, confirming the importance of AF2 in the E2-induced transcriptional activity of ERα. (ii) The uterotrophic action of E2 was totally absent in ERαAF2(0) mice, showing the crucial role of ERαAF2 in E2-induced uterus hyperplasia. (iii) ERαAF2 was dispensable for the accelerative action of E2 on endothelial healing, underlining the functionality of ERαAF2(0) in vivo. (iv) Finally, the atheroprotective effect of E2 was abrogated in ERαAF2(0) LDL-r(-/-) mice. Thus, whereas ERαAF1 and ERαAF2 are both required for the uterotrophic action of E2, we show that only ERαAF2 is necessary for its atheroprotective effect.


Asunto(s)
Aterosclerosis/prevención & control , Endotelio Vascular/patología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Sustancias Protectoras/farmacología , Activación Transcripcional/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aterosclerosis/patología , Peso Corporal/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Reproducibilidad de los Resultados , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología
7.
Haematologica ; 97(8): 1131-5, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22371175

RESUMEN

Hematopoietic stem and progenitor cells reside in vascular and endosteal niches in the bone marrow. Factors affecting bone remodeling were reported to influence numbers and mobilization of hematopoietic stem cells. We therefore analyzed the effects of estradiol acting anabolic on bone integrity. Here we observe that estradiol increases progenitor cell numbers in the vascular but not in the endosteal compartment independent of its estrogen receptor α-dependent anabolic bone effects. Hematopoietic progenitors capable of reconstituting lethally irradiated mice are increased by enhanced cell cycle entry, leading to a diminished long-term reconstitution potential after serial transplantation. We demonstrate that estradiol action on stromal cells potently favors hematopoietic progenitor/stem cell frequency accompanied by enhanced expression of cell adhesion molecules. Finally, estradiol treatment enhances retention of hematopoietic stem cells in the vascular niche of the bone marrow. We describe for the first time the mechanism of estrogen action on hematopoietic stem and progenitor cells.


Asunto(s)
Huesos/efectos de los fármacos , Estradiol/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nicho de Células Madre/efectos de los fármacos
8.
J Immunol ; 185(2): 1169-76, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20554954

RESUMEN

17Beta-estradiol (E2) has been shown to promote the expression of inflammatory mediators by LPS-activated tissue resident macrophages through estrogen receptor alpha (ERalpha) signaling. However, it remained to be determined whether E2 similarly influences macrophages effector functions under inflammatory conditions in vivo, and whether this action of E2 resulted from a direct effect on macrophages. We show in this study that chronic E2 administration to ovariectomized mice significantly increased both cytokine (IL-1beta, IL-6, and TNF-alpha) and inducible NO synthase mRNA abundance in thioglycolate (TGC)-elicited macrophages. The proinflammatory action of E2 was also evidenced at the level of released IL-1beta and IL-6 by ex vivo LPS-activated macrophages. E2 concomitantly inhibited PI3K activity as well as Akt phosphorylation in TGC-elicited macrophages, suggesting that E2 promoted TLR-dependent macrophage activation by alleviating this suppressive signaling pathway. Indeed, this effect was abolished in the presence of the inhibitor wortmannin, demonstrating a key functional link between inhibition of PI3K activity and the E2 action on macrophage functions. Endogenous estrogens levels circulating in ovary-intact mice were sufficient to promote the above described actions. Finally, thanks to a CreLox strategy, targeted disruption of ERalpha gene in macrophages totally abolished the effect of E2 on the expression of inflammatory mediators by both resident and TGC-elicited peritoneal macrophages. In conclusion, we demonstrate that estrogens, through the activation of ERalpha in macrophages in vivo, enhance their ability to produce inflammatory mediators and cytokines upon subsequent TLR activation.


Asunto(s)
Citocinas/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Western Blotting , Células Cultivadas , Citocinas/genética , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Citometría de Flujo , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Tioglicolatos/farmacología , Receptor Toll-Like 4/genética
9.
Proc Natl Acad Sci U S A ; 106(6): 2053-8, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19188600

RESUMEN

Full-length 66-kDa estrogen receptor alpha (ERalpha) stimulates target gene transcription through two activation functions (AFs), AF-1 in the N-terminal domain and AF-2 in the ligand binding domain. Another physiologically expressed 46-kDa ERalpha isoform lacks the N-terminal A/B domains and is consequently devoid of AF-1. Previous studies in cultured endothelial cells showed that the N-terminal A/B domain might not be required for estradiol (E2)-elicited NO production. To evaluate the involvement of ERalpha AF-1 in the vasculoprotective actions of E2, we generated a targeted deletion of the ERalpha A/B domain in the mouse. In these ERalphaAF-1(0) mice, both basal endothelial NO production and reendothelialization process were increased by E2 administration to a similar extent than in control mice. Furthermore, exogenous E2 similarly decreased fatty streak deposits at the aortic root from both ovariectomized 18-week-old ERalphaAF-1(+/+) LDLr(-/-) (low-density lipoprotein receptor) and ERalphaAF-1(0) LDLr (-/-) mice fed with a hypercholesterolemic diet. In addition, quantification of lesion size on en face preparations of the aortic tree of 8-month-old ovariectomized or intact female mice revealed that ERalpha AF-1 is dispensable for the atheroprotective action of endogenous estrogens. We conclude that ERalpha AF-1 is not required for three major vasculoprotective actions of E2, whereas it is necessary for the effects of E2 on its reproductive targets. Thus, selective ER modulators stimulating ERalpha with minimal activation of ERalpha AF-1 could retain beneficial vascular actions, while minimizing the sexual effects.


Asunto(s)
Estradiol/farmacología , Receptor alfa de Estrógeno/fisiología , Animales , Aterosclerosis/etiología , Células Endoteliales/metabolismo , Receptor alfa de Estrógeno/química , Femenino , Ratones , Ratones Noqueados , Óxido Nítrico/biosíntesis , Ovariectomía , Sustancias Protectoras , Estructura Terciaria de Proteína
10.
Circ Res ; 104(2): 245-54, 12p following 254, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19059842

RESUMEN

Although 17beta-estradiol (E2) is protective in experimental models of myocardial and brain ischemia, its effect on skin ischemia remains unknown. Here, we assessed the protective effect of E2 in a mouse model of skin ischemia, mimicking the surgery of skin flaps. Whereas necrosis appeared in the half portion of the skin flap within 1 week after surgery in ovariectomized mice, it was reduced up to 10-fold when mice were pretreated with E2, at least 3 days before the surgery. The beneficial effect of E2 appeared to be attributable to an increase in skin survival, revealed by measuring viability of ex vivo explants and enhancement of the antiapoptotic Bcl-2 protein expression in vivo. This protective effect on the skin contributed to the protection of the vascular network and facilitated reperfusion, which was found to be accelerated in ovariectomized E2-treated mice, whereas hemorrhages were observed in untreated mice. E2 also increased expression of fibroblast growth factor-2 isoforms in the skin and circulating vascular endothelial growth factor in the serum. Finally, this protective effect of E2 was abolished in estrogen receptor-deficient mice (ERalpha(-/-)) but maintained in chimeric mice reconstituted with ERalpha-deficient bone marrow, indicating dispensable action of E2 in bone marrow-derived cells. This protective effect of E2 was mimicked by treatment with tamoxifen, a selective estrogen receptor modulator. In conclusion, we have demonstrated for the first time that E2 exerts a major preventive effect of skin flap necrosis through a prevention of ischemic-induced skin lesions, including those of the vascular network, which contributes to accelerate the reperfusion of the skin flap.


Asunto(s)
Estradiol/metabolismo , Isquemia/prevención & control , Piel/irrigación sanguínea , Piel/metabolismo , Colgajos Quirúrgicos/efectos adversos , Animales , Trasplante de Médula Ósea , Supervivencia Celular , Procedimientos Quirúrgicos Dermatologicos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Isquemia/etiología , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Ratones , Ratones Pelados , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Necrosis , Ovariectomía , Flujo Sanguíneo Regional , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Piel/patología , Tamoxifeno/farmacología , Factores de Tiempo
11.
Proc Natl Acad Sci U S A ; 105(7): 2433-8, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18268329

RESUMEN

Estrogen signaling is mediated by estrogen receptors alpha (ERalpha) and beta (ERbeta). Although a consensus has now been reached concerning many physiological functions of ERalpha, those of ERbeta are still controversial: When housed and examined in two distant laboratories, mice originating from the same initial ERbeta mutant exhibited widely different phenotypes, which were themselves different from the phenotype of another ERbeta mutant previously generated in our laboratory. Because, in addition to a knockout insertion in exon 3, all these mouse mutants displayed alternative splicing transcripts, we have now constructed a ERbeta mouse mutant (ERbeta(ST)(L-/L-)) in which exon 3 was cleanly deleted by Cre/LoxP-mediated excision and was devoid of any transcript downstream of exon 3. Both females and males were sterile. The histology of the ovary was mildly affected, and no histological defects were detected in other organs, neither in females nor in males. Our present results, which are in contrast with previously published data, suggest that, with the notable exception of male and female reproduction, ERbeta is not required in the mouse for the development and homeostasis of the major body systems.


Asunto(s)
Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/metabolismo , Infertilidad/metabolismo , Infertilidad/patología , Neuronas/metabolismo , Animales , Apoptosis , Forma de la Célula , Receptor beta de Estrógeno/genética , Femenino , Infertilidad/genética , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Especificidad de Órganos , Próstata/citología , Próstata/metabolismo
12.
J Pharmacol Exp Ther ; 328(1): 174-82, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18832649

RESUMEN

Estrogen is known to affect vascular function and diabetes development, but the relative contribution of estrogen receptor (ER) isoforms is unclear. The aim of this study was to determine how individual ER isoforms modulate inflammatory enzymes in the vascular wall of control and streptozotocin (STZ)-injected rodents. Primary cultures of rat aortic smooth muscle cells (SMCs) were stimulated with inflammatory agents in the presence or absence of increasing concentrations of the ER alpha and ER beta-selective agonists 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) and diarylpropionitrile (DPN), respectively. The production of inducible nitric-oxide synthase (iNOS), a classical indicator of vascular inflammation, was significantly reduced by PPT in control but not diabetic SMCs, whereas it was further enhanced by DPN treatment in both groups. This distinct action profile was not related to changes in ER transcriptional activity. However, extracellular signal-regulated kinase 1/2 signaling was activated by DPN but not by PPT in cytokine-treated SMCs. In cultured aortic rings from both normoglycemic and STZ-diabetic mice, pharmacological activation of ER alpha attenuated cytokine-driven iNOS induction by 30 to 50%. Vascular iNOS levels were decreased consistently when adding 1 nM 17beta-estradiol to aortic tissues from ER beta- but not ER alpha-knockout mice. These findings suggest a possible role for ER alpha-selective ligands in reducing vascular inflammatory responses under normo- and hyperglycemic conditions.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/fisiología , Músculo Liso Vascular/fisiopatología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Aorta/enzimología , Aorta/fisiopatología , Diabetes Mellitus Experimental/enzimología , Inducción Enzimática , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Inflamación/enzimología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Nitrilos/farmacología , Nitritos/metabolismo , Fenoles , Propionatos/farmacología , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Transfección
13.
J Neurosci ; 27(40): 10832-9, 2007 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17913916

RESUMEN

Learning motor skills is critical for motor abilities such as driving a car or playing piano. The speed at which we learn those skills is subject to many factors. Yet, it is not known to what extent gonadal hormones can affect the achievement of accurate movements in time and space. Here we demonstrate via different lines of evidence that estradiol promotes plasticity in the cerebellar cortex underlying motor learning. First, we show that estradiol enhances induction of long-term potentiation at the parallel fiber to Purkinje cell synapse, whereas it does not affect long-term depression; second, we show that estradiol activation of estrogen receptor beta receptors in Purkinje cells significantly improves gain-decrease adaptation of the vestibulo-ocular reflex, whereas it does not affect general eye movement performance; and third, we show that estradiol increases the density of parallel fiber to Purkinje cell synapses, whereas it does not affect the density of climbing fiber synapses. We conclude that estradiol can improve motor skills by potentiating cerebellar plasticity and synapse formation. These processes may be advantageous during periods of high estradiol levels of the estrous cycle or pregnancy.


Asunto(s)
Cerebelo/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Memoria/efectos de los fármacos , Células de Purkinje/efectos de los fármacos , Análisis de Varianza , Animales , Conducta Animal , Cerebelo/citología , Cerebelo/fisiología , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Receptor beta de Estrógeno/deficiencia , Femenino , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Potenciación a Largo Plazo/efectos de la radiación , Masculino , Memoria/fisiología , Ratones , Ratones Noqueados , Actividad Motora/genética , Fibras Nerviosas/fisiología , Fibras Nerviosas/efectos de la radiación , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Inhibición Neural/efectos de la radiación , Ovariectomía/métodos , Técnicas de Placa-Clamp/métodos , Células de Purkinje/fisiología , Células de Purkinje/ultraestructura , Reflejo Vestibuloocular/fisiología , Factores de Tiempo
14.
Endocrinology ; 149(1): 20-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17947360

RESUMEN

Estrogens play a central role in regulating female reproduction throughout the reproductive axis, and the pituitary is one of the major targets of estrogen action. We hypothesized that estrogen receptor alpha (ERalpha) mediates estrogen action in the pituitary gonadotroph. To test this hypothesis, we generated a mouse line with a selective ERalpha deletion in the gonadotropin alpha-subunit (alphaGSU)-expressing pituitary cells (pituitary-specific ERalpha knockout; ERalpha(flox/flox) alphaGSU(cre)). Although the ERalpha(flox/flox) alphaGSU(cre) female mice maintain a basal level of serum LH and FSH and their ovulatory capacity is comparable to that in controls, they do not display regular estrous cycles and are infertile, indicating a potential disorder in regulating LH and/or FSH secretion. The ERalpha(flox/flox) alphaGSU(cre) female mice express equivalent levels of LHbeta and alphaGSU mRNA compared with wild-type mice as determined by microarray analysis. Taken together, these findings indicate that pituitary gonadotroph ERalpha carries out the effects of estrogens with regard to estrous cyclicity and ultimately fertility.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Fertilidad/genética , Gonadotrofos/metabolismo , Animales , Gonadotropina Coriónica/genética , Gonadotropina Coriónica/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Ciclo Estral/efectos de los fármacos , Ciclo Estral/genética , Ciclo Estral/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante de Subunidad beta/genética , Regulación de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Hormonas Glicoproteicas de Subunidad alfa/metabolismo , Hormona Luteinizante/metabolismo , Hormona Luteinizante de Subunidad beta/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo
15.
Biochem Biophys Res Commun ; 371(2): 320-3, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18439911

RESUMEN

Diabetic Akita male mice are more hyperphagic because of downregulation of proopiomelanocortin (POMC) caused by hypoleptinemia. We investigated the role of estrogen receptor alpha (ERalpha) in the regulation of the hypothalamic POMC in females. ERaKOAkt mice consumed 30% greater food (g/3 weeks) than the Akita diabetic controls. Ovariectomized diabetic (AFO) and nondiabetic (B6FO) mice had significantly lower food intake and elevated serum leptin levels. ERaKOAkt and ERaKO mice also increased serum leptin concentrations, while hypoinsulinemia was observed in ERaKOAkt and hyperinsulinemia in ERaKO mice. RT-PCR showed a significant attenuation of POMC expression in both ERaKOAkt and ERaKO mice, irrespective of the elevated leptin serum levels or hyperinsulinemia, while elevated serum leptin levels in AFO and B6FO mice upregulated POMC gene expression. These results indicate that ERalpha plays an essential role in leptin- and insulin-stimulated upregulation of the POMC gene. This action of ERalpha is likely mediated in a ligand-independent manner.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Regulación de la Expresión Génica , Hiperfagia/genética , Insulina/metabolismo , Leptina/metabolismo , Proopiomelanocortina/genética , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/genética , Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Insulina/sangre , Insulina/farmacología , Leptina/sangre , Leptina/farmacología , Ratones , Ratones Noqueados , Regulación hacia Arriba
16.
Nat Commun ; 9(1): 4723, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413705

RESUMEN

Oestrogen receptor α (ERα) is a transcription factor with ligand-independent and ligand-dependent activation functions (AF)-1 and -2. Oestrogens control postnatal mammary gland development acting on a subset of mammary epithelial cells (MECs), termed sensor cells, which are ERα-positive by immunohistochemistry (IHC) and secrete paracrine factors, which stimulate ERα-negative responder cells. Here we show that deletion of AF-1 or AF-2 blocks pubertal ductal growth and subsequent development because both are required for expression of essential paracrine mediators. Thirty percent of the luminal cells are ERα-negative by IHC but express Esr1 transcripts. This low level ERα expression through AF-2 is essential for cell expansion during puberty and growth-inhibitory during pregnancy. Cell-intrinsic ERα is not required for cell proliferation nor for secretory differentiation but controls transcript levels of cell motility and cell adhesion genes and a stem cell and epithelial mesenchymal transition (EMT) signature identifying ERα as a key regulator of mammary epithelial cell plasticity.


Asunto(s)
Epitelio/metabolismo , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Glándulas Mamarias Animales/metabolismo , Animales , Proliferación Celular , Sistema Endocrino/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Regulación de la Expresión Génica , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esteroides/metabolismo , Relación Estructura-Actividad
17.
J Neurosci ; 26(21): 5649-55, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16723521

RESUMEN

Estrogens are involved in the hypothalamic control of multiple homeostatic functions including reproduction, stress responses, energy metabolism, sleep cycles, temperature regulation, and motivated behaviors. The critical role of 17beta-estradiol (E2) is evident in hypoestrogenic states (e.g., postmenopause) in which many of these functions go awry. The actions of E2 in the brain have been attributed to the activation of estrogen receptors alpha and beta through nuclear, cytoplasmic, or membrane actions. However, we have identified a putative membrane-associated estrogen receptor that is coupled to desensitization of GABAB and mu-opioid receptors in guinea pig and mouse hypothalamic proopiomelanocortin neurons. We have synthesized a new nonsteroidal compound, STX, which selectively targets the Galphaq-coupled phospholipase C-protein kinase C-protein kinase A pathway, and have established that STX is more potent than E2 in mediating this desensitization in an ICI 182, 780-sensitive manner in both guinea pig and mouse neurons. Both E2 and STX were fully efficacious in estrogen receptor alpha,beta knock-out mice. Moreover, in vivo treatment with STX, similar to E2, attenuated the weight gain in hypoestrogenic female guinea pigs. Therefore, this membrane-delimited signaling pathway plays a critical role in the control of energy homeostasis and may provide a novel therapeutic target for treatment of postmenopausal symptoms and eating disorders in females.


Asunto(s)
Peso Corporal/fisiología , Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/fisiología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Femenino , Cobayas , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
18.
Endocrinology ; 148(4): 1902-10, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17204553

RESUMEN

The role of estrogen signaling in the male skeleton via estrogen receptor (ER)-alpha is now well established. ERalpha can elicit responses through either classical estrogen response elements (ERE) pathways or nonclassical, non-ERE pathways. In the present study, we examined the effects of either the attenuation or loss of classical ERalpha signaling on the murine male skeleton. To accomplish this, we crossed male mice heterozygous for a knock-in mutation [nonclassical ERalpha knock-in (NERKI)], which abolishes the ERE-mediated pathway with female heterozygous ERalpha knockout mice (ERalpha+/-) and studied the F1 generation ERalpha+/+, ERalpha+/-, ERalpha+/NERKI, and ERalpha-/NERKI male progeny longitudinally using bone density and histomorphometry. The only ERalpha allele present in ERalpha-/NERKI mice is incapable of classical ERE-mediated signaling, whereas the heterozygous ERalpha+/NERKI mice have both one intact ERalpha and one NERKI allele. As compared with ERalpha+/+ littermates (n=10/genotype), male ERalpha+/NERKI and ERalpha-/NERKI mice displayed axial and appendicular skeletal osteopenia at 6, 12, 20, and 25 wk of age, as demonstrated by significant reductions in total bone mineral density (BMD) at representative sites (areal BMD by dual-energy x-ray absorptiometry at the lumbar vertebrae and femur and volumetric BMD by peripheral quantitative computed tomography at the tibia; P<0.05-0.001 vs. ERalpha+/+). The observed osteopenia in these mice was evident in both trabecular and cortical bone compartments. However, these decreases were more severe in mice lacking classical ERalpha signaling (ERalpha-/NERKI mice), compared with mice in which one wild-type ERalpha allele was present (ERalpha+/NERKI mice). Collectively, these data demonstrate that classical ERalpha signaling is crucial for the development of the murine male skeleton.


Asunto(s)
Huesos/anatomía & histología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Elementos de Respuesta/fisiología , Animales , Peso Corporal , Densidad Ósea , Desarrollo Óseo/genética , Huesos/metabolismo , Fuerza Compresiva , Femenino , Vértebras Lumbares/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
19.
Circulation ; 111(18): 2282-90, 2005 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15867180

RESUMEN

BACKGROUND: Major gender-based differences in the incidence of ventricular tachyarrhythmia after myocardial infarction have been shown in humans. Although the underlying mechanisms are unclear, earlier studies suggest that estrogen receptor-mediated effects play a major role in this process. METHODS AND RESULTS: We examined the effect of estrogen receptor alpha (ERalpha) and estrogen receptor beta (ERbeta) on the electrophysiological phenotype in female mice with and without chronic anterior myocardial infarction. There was no significant difference in overall mortality, infarct size, and parameters of left ventricular remodeling when we compared infarcted ERalpha-deficient and ERbeta-deficient mice with infarcted wild-type animals. In the 12-hour telemetric ECG recording 6 weeks after myocardial infarction, surface ECG parameters did not show significant differences in comparisons of ERalpha-deficient mice versus wild-type controls, infarcted versus noninfarcted ERalpha-deficient mice, and infarcted ERalpha-deficient versus infarcted wild-type mice. However, infarcted ERbeta-deficient versus noninfarcted ERbeta-deficient mice showed a significant prolongation of the QT (61+/-6 versus 48+/-8 ms; P<0.05) and QTc intervals (61+/-7 versus 51+/-9 ms; P<0.05) and the JT (42+/-6 versus 31+/-4 ms; P<0.05) and JTc intervals (42+/-7 versus 33+/-4 ms; P<0.05). Furthermore, infarcted ERbeta-deficient versus infarcted wild-type mice showed a significant prolongation of the QT (61+/-6 versus 53+/-8 ms; P<0.05) and QTc intervals (61+/-7 versus 53+/-7 ms; P<0.05) and the JT (42+/-6 versus 31+/-5 ms; P<0.05) and JTc intervals (42+/-7 versus 31+/-5 ms; P<0.05), accompanied by a significant decrease of ventricular premature beats (7+/-21/h versus 71+/-110/h; P<0.05). Finally, real-time polymerase chain reaction-based quantitative analysis of mRNA levels showed a significantly lower expression of Kv4.3 (coding for I(to)) in ERbeta-deficient mice (P<0.05). CONCLUSIONS: Estrogen receptor beta deficiency results in prolonged ventricular repolarization and decreased ventricular automaticity in female mice with chronic myocardial infarction.


Asunto(s)
Electrocardiografía , Receptor beta de Estrógeno/deficiencia , Infarto del Miocardio/complicaciones , Taquicardia Ventricular/etiología , Animales , Receptor alfa de Estrógeno/deficiencia , Femenino , Síndrome de QT Prolongado/etiología , Potenciales de la Membrana , Ratones , Ratones Noqueados , Infarto del Miocardio/fisiopatología , ARN Mensajero/análisis , Canales de Potasio Shal/genética , Complejos Prematuros Ventriculares/etiología
20.
Circ Res ; 90(10): 1087-92, 2002 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-12039798

RESUMEN

Blood vessel cells express the 2 known estrogen receptors, alpha and beta (ERalpha, ERbeta), which are thought to mediate estrogen inhibition of vascular injury and atherosclerosis, but the relative role of ERalpha and ERbeta in these events is controversial. Estrogen inhibits the vascular injury response to the same extent in ovariectomized female wild-type mice and in the original single gene knockout mice for ERalpha (ERalphaKO(Chapel Hill) [ERalphaKO(CH)]) and ERbeta (ERbetaKO(Chapel Hill) [ERbetaKO(CH)]). In double gene knockout mice generated by crossing these animals (ERalpha,betaKO(CH)), estrogen no longer inhibits medial thickening after vascular injury, but still inhibits vascular smooth muscle cell proliferation and increases uterine weight. The partial retention of estrogen responsiveness in ERalpha,betaKO(CH) mice could be due either to the presence of a novel, unidentified estrogen receptor or to functional expression of an estrogen receptor-alpha splice variant in the parental ERalphaKO(CH) mice. To distinguish between these possibilities, we studied recently generated mice fully null for estrogen receptor alpha (ERalphaKO(Strasbourg) [ERalphaKO(St)]) and examined the effect of estrogen on the response to vascular injury. In the present study, we show that after vascular injury in ovariectomized ERalphaKO(St) mice, estrogen has no detectable effect on any measure of vascular injury, including medial area, proteoglycan deposition, or smooth muscle cell proliferation. These data demonstrate that estrogen receptor-alpha mediates the protective effects of estrogen on the response to vascular injury.


Asunto(s)
Enfermedades de las Arterias Carótidas/prevención & control , Estradiol/farmacología , Receptores de Estrógenos/fisiología , Animales , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , División Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , Estradiol/uso terapéutico , Receptor alfa de Estrógeno , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Tamaño de los Órganos , Proteoglicanos/biosíntesis , Receptores de Estrógenos/genética , Útero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA