Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Elife ; 132024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39007235

RESUMEN

The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor long form (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determined the role of glutamatergic neurotransmission from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces luteinizing hormone (LH) release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LeprCre (LepRb-Cre) mice. We collected blood sequentially before and for 1 hr after intravenous clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of Fos immunoreactive neurons in the PMv. Next, females with deletion of Slc17a6 (Vglut2) in LepRb neurons (LeprΔVGlut2) showed delayed age of puberty, disrupted estrous cycles, increased gonadotropin-releasing hormone (GnRH) concentration in the axon terminals, and disrupted LH secretion, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LeprloxTB) with concomitant deletion of Slc17a6 (Vglut2flox) mice. Rescue of Lepr and deletion of Slc17a6 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LeprloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation, and became pregnant, while LeprloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic neurotransmission from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.


Asunto(s)
Ácido Glutámico , Receptores de Leptina , Maduración Sexual , Transmisión Sináptica , Animales , Femenino , Receptores de Leptina/metabolismo , Receptores de Leptina/genética , Ratones , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Reproducción , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Hormona Luteinizante/sangre , Hormona Luteinizante/metabolismo
2.
bioRxiv ; 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-37790549

RESUMEN

The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determine the role of glutamatergic signaling from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces LH release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LepRb-Cre mice. We collected blood sequentially before and for 1h after iv. clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of cFos immunoreactive neurons in the PMv. Next, females with deletion of Vglut2 in LepRb neurons (LepR∆VGlut2) showed delayed age of puberty, disrupted estrous cycles, increased GnRH concentration in the axon terminals and disrupted LH responses, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LepRloxTB) with concomitant deletion of Vglut2 (Vglut2-floxed) mice. Rescue of Lepr and deletion of Vglut2 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LepRloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation and became pregnant, while LepRloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic signaling from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.

3.
Endocrinology ; 159(4): 1718-1733, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29438518

RESUMEN

Obese women are at high risk of pregnancy complications, including preeclampsia, miscarriage, preterm birth, stillbirth, and neonatal death. In the current study, we aimed to determine the effects of obesity on pregnancy outcome and placental gene expression in preclinical mouse models of genetic and nutritional obesity. The leptin receptor (LepR) null-reactivatable (LepRloxTB), LepR-deficient (Leprdb/+), and high-fat diet (HFD)-fed mice were assessed for fertility, pregnancy outcome, placental morphology, and placental transcriptome using standard quantitative polymerase chain reaction (qPCR) and qPCR arrays. The restoration of fertility of LepRloxTB was performed by stereotaxic delivery of adeno-associated virus-Cre into the hypothalamic ventral premammillary nucleus. Fertile LepRloxTB females were morbidly obese, whereas the wild-type mice-fed HFD showed only a mild increase in body weight. Approximately 80% of the LepRloxTB females had embryo resorptions (∼40% of the embryos). In HFD mice, the number of resorptions was not different from controls fed a regular diet. Placentas of resorbed embryos from obese mice displayed necrosis and inflammatory infiltrate in the labyrinth and changes in the expression of genes associated with angiogenesis and inflammation (e.g., Vegfa, Hif1a, Nfkbia, Tlr3, Tlr4). In contrast, placentas from embryos of females on HFD showed changes in a different set of genes, mostly associated with cellular growth and response to stress (e.g., Plg, Ang, Igf1, Igfbp1, Fgf2, Tgfb2, Serpinf1). Sexual dimorphism in gene expression was only apparent in placentas from obese LepRloxTB mice. Our findings indicate that an obese environment and HFD have distinct effects on pregnancy outcome and the placental transcriptome.


Asunto(s)
Dieta Alta en Grasa , Regulación de la Expresión Génica , Obesidad/genética , Placenta/metabolismo , Receptores de Leptina/genética , Animales , Femenino , Hipotálamo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Leptina/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Obesidad/metabolismo , Embarazo , Resultado del Embarazo , Receptores de Leptina/metabolismo
4.
JCI Insight ; 2(23)2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29212950

RESUMEN

The role of PI3K in leptin physiology has been difficult to determine due to its actions downstream of several metabolic cues, including insulin. Here, we used a series of mouse models to dissociate the roles of specific PI3K catalytic subunits and of insulin receptor (InsR) downstream of leptin signaling. We show that disruption of p110α and p110ß subunits in leptin receptor cells (LRΔα+ß) produces a lean phenotype associated with increased energy expenditure, locomotor activity, and thermogenesis. LRΔα+ß mice have deficient growth and delayed puberty. Single subunit deletion (i.e., p110α in LRΔα) resulted in similarly increased energy expenditure, deficient growth, and pubertal development, but LRΔα mice have normal locomotor activity and thermogenesis. Blunted PI3K in leptin receptor (LR) cells enhanced leptin sensitivity in metabolic regulation due to increased basal hypothalamic pAKT, leptin-induced pSTAT3, and decreased PTEN levels. However, these mice are unresponsive to leptin's effects on growth and puberty. We further assessed if these phenotypes were associated with disruption of insulin signaling. LRΔInsR mice have no metabolic or growth deficit and show only mild delay in pubertal completion. Our findings demonstrate that PI3K in LR cells plays an essential role in energy expenditure, growth, and reproduction. These actions are independent from insulin signaling.


Asunto(s)
Leptina/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Receptores de Leptina/metabolismo , Animales , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Estro/fisiología , Femenino , Fertilidad/fisiología , Eliminación de Gen , Silenciador del Gen , Crecimiento/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Pubertad/fisiología , Receptor de Insulina/deficiencia , Receptor de Insulina/fisiología , Maduración Sexual/fisiología , Transducción de Señal/fisiología
5.
Brain Struct Funct ; 222(9): 4111-4129, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28616754

RESUMEN

Prokineticin receptor 2 (PROKR2) is predominantly expressed in the mammalian central nervous system. Loss-of-function mutations of PROKR2 in humans are associated with Kallmann syndrome due to the disruption of gonadotropin releasing hormone neuronal migration and deficient olfactory bulb morphogenesis. PROKR2 has been also implicated in the neuroendocrine control of GnRH neurons post-migration and other physiological systems. However, the brain circuitry and mechanisms associated with these actions have been difficult to investigate mainly due to the widespread distribution of Prokr2-expressing cells, and the lack of animal models and molecular tools. Here, we describe the generation, validation and characterization of a new mouse model that expresses Cre recombinase driven by the Prokr2 promoter, using CRISPR-Cas9 technology. Cre expression was visualized using reporter genes, tdTomato and GFP, in males and females. Expression of Cre-induced reporter genes was found in brain sites previously described to express Prokr2, e.g., the paraventricular and the suprachiasmatic nuclei, and the area postrema. The Prokr2-Cre mouse model was further validated by colocalization of Cre-induced GFP and Prokr2 mRNA. No disruption of Prokr2 expression, GnRH neuronal migration or fertility was observed. Comparative analysis of Prokr2-Cre expression in male and female brains revealed a sexually dimorphic distribution confirmed by in situ hybridization. In females, higher Cre activity was found in the medial preoptic area, ventromedial nucleus of the hypothalamus, arcuate nucleus, medial amygdala and lateral parabrachial nucleus. In males, Cre was higher in the amygdalo-hippocampal area. The sexually dimorphic pattern of Prokr2 expression indicates differential roles in reproductive function and, potentially, in other physiological systems.


Asunto(s)
Encéfalo/patología , Síndrome de Kallmann/patología , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Caracteres Sexuales , Animales , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrasas , Síndrome de Kallmann/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo
6.
PLoS One ; 9(8): e103775, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25090643

RESUMEN

BACKGROUND: Anorexia nervosa is a primary psychiatric disorder, with non-negligible rates of mortality and morbidity. Some of the related alterations could participate in a vicious cycle limiting the recovery. Animal models mimicking various physiological alterations related to anorexia nervosa are necessary to provide better strategies of treatment. AIM: To explore physiological alterations and recovery in a long-term mouse model mimicking numerous consequences of severe anorexia nervosa. METHODS: C57Bl/6 female mice were submitted to a separation-based anorexia protocol combining separation and time-restricted feeding for 10 weeks. Thereafter, mice were housed in standard conditions for 10 weeks. Body weight, food intake, body composition, plasma levels of leptin, adiponectin, IGF-1, blood levels of GH, reproductive function and glucose tolerance were followed. Gene expression of several markers of lipid and energy metabolism was assayed in adipose tissues. RESULTS: Mimicking what is observed in anorexia nervosa patients, and despite a food intake close to that of control mice, separation-based anorexia mice displayed marked alterations in body weight, fat mass, lean mass, bone mass acquisition, reproductive function, GH/IGF-1 axis, and leptinemia. mRNA levels of markers of lipogenesis, lipolysis, and the brown-like adipocyte lineage in subcutaneous adipose tissue were also changed. All these alterations were corrected during the recovery phase, except for the hypoleptinemia that persisted despite the full recovery of fat mass. CONCLUSION: This study strongly supports the separation-based anorexia protocol as a valuable model of long-term negative energy balance state that closely mimics various symptoms observed in anorexia nervosa, including metabolic adaptations. Interestingly, during a recovery phase, mice showed a high capacity to normalize these parameters with the exception of plasma leptin levels. It will be interesting therefore to explore further the central and peripheral effects of the uncorrected hypoleptinemia during recovery from separation-based anorexia.


Asunto(s)
Anorexia Nerviosa/fisiopatología , Ansiedad de Separación/fisiopatología , Conducta Alimentaria , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Adiponectina/metabolismo , Tejido Adiposo/patología , Animales , Anorexia Nerviosa/sangre , Anorexia Nerviosa/complicaciones , Anorexia Nerviosa/genética , Ansiedad de Separación/sangre , Ansiedad de Separación/complicaciones , Composición Corporal , Modelos Animales de Enfermedad , Ingestión de Alimentos , Ciclo Estral , Femenino , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leptina/sangre , Metabolismo de los Lípidos/genética , Ratones Endogámicos C57BL , Oxidación-Reducción , Fenotipo , Reproducción , Factores de Tiempo , Pérdida de Peso
7.
J Clin Invest ; 124(6): 2550-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24812663

RESUMEN

The transition to puberty and adult fertility both require a minimum level of energy availability. The adipocyte-derived hormone leptin signals the long-term status of peripheral energy stores and serves as a key metabolic messenger to the neuroendocrine reproductive axis. Humans and mice lacking leptin or its receptor fail to complete puberty and are infertile. Restoration of leptin levels in these individuals promotes sexual maturation, which requires the pulsatile, coordinated delivery of gonadotropin-releasing hormone to the pituitary and the resulting surge of luteinizing hormone (LH); however, the neural circuits that control the leptin-mediated induction of the reproductive axis are not fully understood. Here, we found that leptin coordinated fertility by acting on neurons in the preoptic region of the hypothalamus and inducing the synthesis of the freely diffusible volume-based transmitter NO, through the activation of neuronal NO synthase (nNOS) in these neurons. The deletion of the gene encoding nNOS or its pharmacological inhibition in the preoptic region blunted the stimulatory action of exogenous leptin on LH secretion and prevented the restoration of fertility in leptin-deficient female mice by leptin treatment. Together, these data indicate that leptin plays a central role in regulating the hypothalamo-pituitary-gonadal axis in vivo through the activation of nNOS in neurons of the preoptic region.


Asunto(s)
Leptina/metabolismo , Óxido Nítrico/metabolismo , Área Preóptica/fisiología , Reproducción/fisiología , Animales , Femenino , Humanos , Kisspeptinas/metabolismo , Leptina/deficiencia , Leptina/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Área Preóptica/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Transducción de Señal
8.
Horm Behav ; 66(1): 7-14, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24746731

RESUMEN

This article is part of a Special Issue Energy Balance. Over the past two decades, and in particular over the past 5-7 years, there has been a tremendous advancement in the understanding of the metabolic control of reproductive physiology. This has been in large part due to the advancement and refinement of gene targeting tools and techniques for molecular mapping. Yet despite the emergence of exciting and often times thought-provoking data through the use of new mouse models, the heavy reliance on gene targeting strategies has become fundamental in this process and thus caution must be exercised when interpreting results. This minireview article will explore the generation of new mouse models using genetic manipulation, such as viral vector delivery and the use of the Cre/loxP system, to investigate the role of circulating metabolic hormones in the coordination of reproductive physiology. In addition, we will also highlight some of the pitfalls in the use of genetic manipulation in the current paradigms. However, it has become clear that metabolic cues employ integrated and plastic neural circuits in order to modulate the neuroendocrine reproductive axis, and despite recent advances much remains to be elucidated about this circuitry.


Asunto(s)
Ghrelina/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Ratones Transgénicos , Reproducción/fisiología , Transducción de Señal/fisiología , Animales , Ratones
9.
J Neurosci ; 32(3): 932-45, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22262891

RESUMEN

Reproduction is controlled in the brain by a neural network that drives the secretion of gonadotropin-releasing hormone (GnRH). Various permissive homeostatic signals must be integrated to achieve ovulation in mammals. However, the neural events controlling the timely activation of GnRH neurons are not completely understood. Here we show that kisspeptin, a potent activator of GnRH neuronal activity, directly communicates with neurons that synthesize the gaseous transmitter nitric oxide (NO) in the preoptic region to coordinate the progression of the ovarian cycle. Using a transgenic Gpr54-null IRES-LacZ knock-in mouse model, we demonstrate that neurons containing neuronal NO synthase (nNOS), which are morphologically associated with kisspeptin fibers, express the kisspeptin receptor GPR54 in the preoptic region, but not in the tuberal region of the hypothalamus. The activation of kisspeptin signaling in preoptic neurons promotes the activation of nNOS through its phosphorylation on serine 1412 via the AKT pathway and mimics the positive feedback effects of estrogens. Finally, we show that while NO release restrains the reproductive axis at stages of the ovarian cycle during which estrogens exert their inhibitory feedback, it is required for the kisspeptin-dependent preovulatory activation of GnRH neurons. Thus, interactions between kisspeptin and nNOS neurons may play a central role in regulating the hypothalamic-pituitary-gonadal axis in vivo.


Asunto(s)
Hipotálamo/citología , Kisspeptinas/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Ovulación/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Ciclo Estral/efectos de los fármacos , Ciclo Estral/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipotálamo/efectos de los fármacos , Kisspeptinas/deficiencia , Kisspeptinas/farmacología , Hormona Luteinizante/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/deficiencia , Ovulación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Kisspeptina-1 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esteroides/farmacología
10.
Neuroendocrinology ; 93(2): 74-89, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21335953

RESUMEN

Nitric oxide (NO) is a peculiar chemical transmitter that freely diffuses through aqueous and lipid environments and plays a role in major aspects of brain function. Within the hypothalamus, NO exerts critical effects upon the gonadotropin-releasing hormone (GnRH) network to maintain fertility. Here, we review recent evidence that NO regulates major aspects of the GnRH neuron physiology. Far more active than once thought, NO powerfully controls GnRH neuronal activity, GnRH release and structural plasticity at the neurohemal junction. In the preoptic region, neuronal nitric oxide synthase (nNOS) activity is tightly regulated by estrogens and is found to be maximal at the proestrus stage. Natural fluctuations of estrogens control both the differential coupling of this Ca²+-activated enzyme to glutamate N-methyl-D-aspartic acid receptor channels and phosphorylation-mediated nNOS activation. Furthermore, NO endogenously produced by neurons expressing nNOS acutely and directly suppresses spontaneous firing in GnRH neurons, which suggests that neuronal NO may serve as a synchronizing switch within the preoptic region. At the median eminence, NO is spontaneously released from an endothelial source and follows a pulsatile and cyclic pattern of secretion. Importantly, GnRH release appears to be causally related to endothelial NO release. NO is also highly involved in mediating the dialogue set in motion between vascular endothelial cells and tanycytes that control the direct access of GnRH neurons to the pituitary portal blood during the estrous cycle. Altogether, these data raise the intriguing possibility that the neuroendocrine brain uses NO to coordinate both GnRH neuronal activity and GnRH release at key stages of reproductive physiology.


Asunto(s)
Encéfalo/fisiología , Células Endoteliales/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sistemas Neurosecretores/fisiología , Óxido Nítrico/fisiología , Reproducción/fisiología , Transducción de Señal/fisiología , Animales , Hormona Liberadora de Gonadotropina/fisiología , Modelos Biológicos , Óxido Nítrico/biosíntesis
11.
Front Neuroendocrinol ; 31(3): 241-58, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20546773

RESUMEN

As the final common pathway for the central control of gonadotropin secretion, GnRH neurons are subjected to numerous regulatory homeostatic and external factors to achieve levels of fertility appropriate to the organism. The GnRH system thus provides an excellent model in which to investigate the complex relationships between neurosecretion, morphological plasticity and the expression of a physiological function. Throughout the reproductive cycle beginning from postnatal sexual development and the onset of puberty to reproductive senescence, and even within the ovarian cycle itself, all levels of the GnRH system undergo morphological plasticity. This structural plasticity within the GnRH system appears crucial to the timely control of reproductive competence within the individual, and as such must have coordinated actions of multiple signals secreted from glial cells, endothelial cells, and GnRH neurons. Thus, the GnRH system must be viewed as a complete neuro-glial-vascular unit that works in concert to maintain the reproductive axis.


Asunto(s)
Comunicación Celular/fisiología , Células Endoteliales/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuroglía/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Animales , Células Endoteliales/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Humanos , Modelos Biológicos , Neuroglía/metabolismo , Neuronas/metabolismo , Ovario/metabolismo , Ovario/fisiología , Pubertad/metabolismo , Pubertad/fisiología , Receptores LHRH/metabolismo , Receptores LHRH/fisiología
12.
Endocrinology ; 151(6): 2723-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20371700

RESUMEN

Within the preoptic region, nitric oxide (NO) production varies during the ovarian cycle and has the ability to impact hypothalamic reproductive function. One mechanism for the regulation of NO release mediated by estrogens during the estrous cycle includes physical association of the calcium-activated neuronal NO synthase (nNOS) enzyme with the glutamate N-methyl-d-aspartate (NMDA) receptor channels via the postsynaptic density 95 scaffolding protein. Here we demonstrate that endogenous variations in estrogens levels during the estrous cycle also coincide with corresponding changes in the state of nNOS Ser1412 phosphorylation, the level of association of this isoform with the NMDA receptor/postsynaptic density 95 complex at the plasma membrane, and the activity of NO synthase (NOS). Neuronal NOS Ser1412 phosphorylation is maximal on the afternoon of proestrus when both the levels of estrogens and the physical association of nNOS with NMDA receptors are highest. Estradiol mimicked these effects in ovariectomized (OVX) rats. In addition, the catalytic activity of NOS in membrane protein extracts from the preoptic region, i.e. independent of any functional protein-protein interactions or cell-cell signaling, was significantly increased in estradiol-treated OVX rats compared with OVX rats. Finally, lambda phosphatase-mediated nNOS dephosphorylation dramatically impaired NOS activity in preoptic region protein extracts, thus demonstrating the important role of phosphorylation in the regulation of NO production in the preoptic region. Taken together, these results yield new insights into the regulation of neuron-derived NO production by gonadal steroids within the preoptic region and raise the possibility that changes in nNOS phosphorylation during fluctuating physiological conditions may be involved in the hypothalamic control of key neuroendocrine functions, such as reproduction.


Asunto(s)
Estrógenos/metabolismo , Hipotálamo/metabolismo , Ciclo Menstrual/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Western Blotting , Membrana Celular/metabolismo , Estradiol/farmacología , Femenino , Hipotálamo/efectos de los fármacos , Inmunoprecipitación , Ciclo Menstrual/fisiología , Ovariectomía , Fosforilación/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley
13.
Physiol Behav ; 93(3): 588-94, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18037458

RESUMEN

Age of puberty was examined in female mice through non-invasive behavioral and fertility measures, in relationship to ano-genital distance, phytoestrogen content of diet, and exposure to males post-weaning. Throughout gestation and post-natal development, females were exposed to a regular diet or one that was nutritionally similar but deficient in phytoestrogens. After segregation at weaning on the basis of a short or long ano-genital distance index (AGDI), an indirect measure of in utero androgen exposure, females were housed alone or underneath two outbred adult males for two weeks. Subsequently, an outbred male was placed in the cage of each developing female, and mating behavior, escape attempts, biting gestures, and boxing postures were recorded. Next, females were monitored for the occurrence of a copulatory plug and allowed to bear young, with pregnancy and litters monitored up to weaning. Male-exposed females fed a regular diet were immediately sexually receptive when housed directly with males, and their conceptions occurred earlier than did those of other females. Subjects fed a diet deficient in phytoestrogens were least likely to show sexual receptivity. Male-exposed females with longer AGDI displayed more escape attempts in the presence of males, regardless of diet. Once inseminated, most females carried to term and the majority of pups survived until weaning. These data suggest that phytoestrogens and AGDI interact with exposure to males in determining age at onset of puberty.


Asunto(s)
Dieta , Genitales Femeninos/anatomía & histología , Fitoestrógenos/administración & dosificación , Conducta Sexual Animal/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Conducta Sexual Animal/fisiología , Maduración Sexual/fisiología , Factores de Tiempo , Útero/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA