Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Tipo de estudio
Intervalo de año de publicación
1.
Endocrinology ; 154(10): 3836-46, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24025225

RESUMEN

The principal nucleus of the bed nucleus of the stria terminalis (BNSTp) and anteroventral periventricular nucleus of the hypothalamus (AVPV) are sexually dimorphic, hormone-sensitive forebrain regions. Here we report a profound sex difference in estrogen receptor-α (ERα) immunoreactivity (IR) in the BNSTp, with robust ERα IR in females and the near absence of labeling in males. This sex difference is due to the suppression of ERα IR by testicular hormones in adulthood: it was not present at birth and was not altered by neonatal treatment of females with estradiol; gonadectomy of adult males increased ERα IR to that of females, whereas gonadectomy of adult females had no effect. Treating gonadally intact males with an aromatase inhibitor partially feminized ERα IR in the BNSTp, suggesting that testicular suppression required aromatization. By contrast, in AVPV we found a modest sex difference in ERα IR that was relatively insensitive to steroid manipulations in adulthood. ERα IR in AVPV was, however, masculinized in females treated with estradiol at birth, suggesting that the sex difference is due to organizational effects of estrogens. The difference in ERα IR in the BNSTp of males and females appears to be at least in part due to greater expression of mRNA of the ERα gene (Esr1) in females. The sex difference in message is smaller than the difference in immunoreactivity, however, suggesting that posttranscriptional mechanisms also contribute to the pronounced suppression of ERα IR and presumably to functions mediated by ERα in the male BNSTp.


Asunto(s)
Núcleos Talámicos Anteriores/metabolismo , Receptor alfa de Estrógeno/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Núcleos Septales/metabolismo , Andrógenos/farmacología , Animales , Animales Recién Nacidos , Núcleos Talámicos Anteriores/citología , Núcleos Talámicos Anteriores/efectos de los fármacos , Núcleos Talámicos Anteriores/crecimiento & desarrollo , Inhibidores de la Aromatasa/farmacología , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Orquiectomía/efectos adversos , Especificidad de Órganos , Ovariectomía/efectos adversos , ARN Mensajero/metabolismo , Núcleos Septales/citología , Núcleos Septales/efectos de los fármacos , Núcleos Septales/crecimiento & desarrollo , Caracteres Sexuales
2.
Endocrinology ; 154(9): 3273-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825121

RESUMEN

Kisspeptin, encoded by Kiss1, stimulates reproduction. In rodents, one Kiss1 population resides in the hypothalamic anterior ventral periventricular nucleus and neighboring rostral periventricular nucleus (AVPV/PeN). AVPV/PeN Kiss1 neurons are sexually dimorphic (greater in females), yet the mechanisms regulating their development and sexual differentiation remain poorly understood. Neonatal estradiol (E2) normally defeminizes AVPV/PeN kisspeptin neurons, but emerging evidence suggests that developmental E2 may also influence feminization of kisspeptin, although exactly when in development this process occurs is unknown. In addition, the obligatory role of GnRH signaling in governing sexual differentiation of Kiss1 or other sexually dimorphic traits remains untested. Here, we assessed whether AVPV/PeN Kiss1 expression is permanently impaired in adult hpg (no GnRH or E2) or C57BL6 mice under different E2 removal or replacement paradigms. We determined that 1) despite lacking GnRH signaling in development, marked sexual differentiation of Kiss1 still occurs in hpg mice; 2) adult hpg females, who lack lifetime GnRH and E2 exposure, have reduced AVPV/PeN Kiss1 expression compared to wild-type females, even after chronic adulthood E2 treatment; 3) E2 exposure to hpg females during the pubertal period does not rescue their submaximal adult Kiss1 levels; and 4) in C57BL6 females, removal of ovarian E2 before the pubertal or juvenile periods does not impair feminization and maximal adult AVPV/PeN Kiss1 expression nor the ability to generate LH surges, indicating that puberty is not a critical period for Kiss1 development. Thus, sexual differentiation still occurs without GnRH, but GnRH or downstream E2 signaling is needed sometime before juvenile development for complete feminization and maximal Kiss1 expression in adult females.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Diferenciación Sexual , Transducción de Señal , Núcleos Talámicos/metabolismo , Regulación hacia Arriba , Animales , Núcleos Talámicos Anteriores/citología , Núcleos Talámicos Anteriores/efectos de los fármacos , Núcleos Talámicos Anteriores/crecimiento & desarrollo , Núcleos Talámicos Anteriores/metabolismo , Estradiol/farmacología , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Terapia de Reemplazo de Estrógeno , Estrógenos/farmacología , Estrógenos/uso terapéutico , Femenino , Hormona Liberadora de Gonadotropina/genética , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/metabolismo , Hipogonadismo/patología , Núcleos Talámicos Intralaminares/citología , Núcleos Talámicos Intralaminares/efectos de los fármacos , Núcleos Talámicos Intralaminares/crecimiento & desarrollo , Núcleos Talámicos Intralaminares/metabolismo , Kisspeptinas/biosíntesis , Kisspeptinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Ovariectomía/efectos adversos , Diferenciación Sexual/efectos de los fármacos , Desarrollo Sexual/efectos de los fármacos , Núcleos Talámicos/citología , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/crecimiento & desarrollo , Regulación hacia Arriba/efectos de los fármacos
3.
Adv Exp Med Biol ; 784: 221-52, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23550009

RESUMEN

The neuropeptide kisspeptin, encoded by the Kiss1 gene, is required for mammalian puberty and fertility. Examining the development of the kisspeptin system contributes to our understanding of pubertal progression and adult reproduction and sheds light on possible mechanisms underlying the development of reproductive disorders, such as precocious puberty or hypogonadotropic hypogonadism. Recent work, primarily in rodent models, has begun to study the development of kisspeptin neurons and their regulation by sex steroids and other factors at early life stages. In the brain, kisspeptin is predominantly expressed in two areas of the hypothalamus, the anteroventral periventricular nucleus and neighboring periventricular nucleus (pre-optic area in some species) and the arcuate nucleus. Kisspeptin neurons in these two hypothalamic regions are differentially regulated by testosterone and estradiol, both in development and in adulthood, and also display differences in their degree of sexual dimorphism. In this chapter, we discuss what is currently known and not known about the ontogeny, maturation, and sexual differentiation of kisspeptin neurons, as well as their regulation by sex steroids and other factors during development.


Asunto(s)
Núcleos Talámicos Anteriores/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Kisspeptinas/metabolismo , Caracteres Sexuales , Adulto , Animales , Núcleos Talámicos Anteriores/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Hipogonadismo/metabolismo , Masculino , Pubertad/fisiología , Pubertad Precoz/metabolismo , Reproducción/fisiología , Testosterona/metabolismo
4.
Front Neuroendocrinol ; 33(1): 36-44, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21741397

RESUMEN

Sex differences in luteinizing hormone (LH) release patterns are controlled by the hypothalamus, established during the perinatal period and required for fertility. Female mammals exhibit a cyclic surge pattern of LH release, while males show a tonic release pattern. In rodents, the LH surge pattern is dictated by the anteroventral periventricular nucleus (AVPV), an estrogen receptor-rich structure that is larger and more cell-dense in females. Sex differences result from mitochondrial cell death triggered in perinatal males by estradiol derived from aromatization of testosterone. Herein we provide an historical perspective and an update describing evidence that molecules important for cell survival and cell death in the immune system also control these processes in the developing AVPV. We conclude with a new model proposing that development of the female AVPV requires constitutive activation of the Tnfα, Tnf receptor 2, NfκB and Bcl2 pathway that is blocked by induction of Tnf receptor-associated factor 2-inhibiting protein (Traip) in the male.


Asunto(s)
Núcleo Hipotalámico Anterior/crecimiento & desarrollo , Núcleos Talámicos Anteriores/crecimiento & desarrollo , Hormona Luteinizante/metabolismo , FN-kappa B/fisiología , Diferenciación Sexual/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Núcleo Hipotalámico Anterior/fisiología , Núcleos Talámicos Anteriores/fisiología , Muerte Celular , Femenino , Masculino , Mitocondrias , Factor 2 Asociado a Receptor de TNF/antagonistas & inhibidores , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA