Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Neuroendocrinology ; 99(2): 94-107, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24686008

RESUMEN

The importance of the Kiss1 gene in the control of reproductive development is well documented. However, much less is known about the transcriptional regulation of Kiss1 expression in the hypothalamus. Critical for these studies is an accurate identification of the site(s) where Kiss1 transcription is initiated. Employing 5'-RACE PCR, we detected a transcription start site (TSS1) used by the hypothalamus of rats, mice, nonhuman primates and humans to initiate Kiss1 transcription. In rodents, an exon 1 encoding 5'-untranslated sequences is followed by an alternatively spliced second exon, which encodes 5'-untranslated regions of two different lengths and contains the translation initiation codon (ATG). In nonhuman primates and humans, exon 2 is not alternatively spliced. Surprisingly, in rat mediobasal hypothalamus (MBH), but not preoptic area (POA), an additional TSS (TSS2) located upstream from TSS1 generates an exon 1 longer (377 bp) than the TSS1-derived exon 1 (98 bp). The content of TSS1-derived transcripts increased at puberty in the POA and MBH of female rats. It also increased in the MBH after ovariectomy, and this change was prevented by estrogen. In contrast, no such changes in TSS2-derived transcript abundance were detected. Promoter assays showed that the proximal TSS1 promoter is much more active than the putative TSS2 promoter, and that only the TSS1 promoter is regulated by estrogen. These differences appear to be related to the presence of a TATA box and binding sites for transcription factors activating transcription and interacting with estrogen receptor-α in the TSS1, but not TSS2, promoter.


Asunto(s)
Estrógenos/farmacología , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , ARN Mensajero/metabolismo , Maduración Sexual , Sitio de Iniciación de la Transcripción , Transcripción Genética/efectos de los fármacos , Animales , Receptor alfa de Estrógeno/efectos de los fármacos , Terapia de Reemplazo de Estrógeno , Exones/genética , Femenino , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Ovariectomía , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Transcripción Genética/genética
2.
Horm Behav ; 64(2): 175-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23998662

RESUMEN

This article is part of a Special Issue "Puberty and Adolescence". Puberty is a major developmental milestone controlled by the interaction of genetic factors and environmental cues of mostly metabolic and circadian nature. An increased pulsatile release of the decapeptide gonadotropin releasing hormone (GnRH) from hypothalamic neurosecretory neurons is required for both the initiation and progression of the pubertal process. This increase is brought about by coordinated changes that occur in neuronal and glial networks associated with GnRH neurons. These changes ultimately result in increased neuronal and glial stimulatory inputs to the GnRH neuronal network and a reduction of transsynaptic inhibitory influences. While some of the major players controlling pubertal GnRH secretion have been identified using gene-centric approaches, much less is known about the system-wide control of the overall process. Because the pubertal activation of GnRH release involves a diversity of cellular phenotypes, and a myriad of intracellular and cell-to-cell signaling molecules, it appears that the overall process is controlled by a highly coordinated and interactive regulatory system involving hundreds, if not thousands, of gene products. In this article we will discuss emerging evidence suggesting that these genes are arranged as functionally connected networks organized, both internally and across sub-networks, in a hierarchical fashion. According to this concept, the core of these networks is composed of transcriptional regulators that, by directing expression of downstream subordinate genes, provide both stability and coordination to the cellular networks involved in initiating the pubertal process. The integrative response of these gene networks to external inputs is postulated to be coordinated by epigenetic mechanisms.


Asunto(s)
Redes Reguladoras de Genes , Sistemas Neurosecretores/fisiología , Primates/fisiología , Maduración Sexual/genética , Biología de Sistemas/métodos , Animales , Epigénesis Genética/fisiología , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratas
3.
Adv Exp Med Biol ; 784: 363-83, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23550015

RESUMEN

Body energy balance and metabolic signals are important modulators of puberty and reproductive function, so that perturbations of metabolism and energy reserves (ranging from persistent energy insufficiency to morbid obesity) are frequently linked to reproductive disorders. The mechanisms for the tight association between body metabolic state and reproduction are multifaceted, and likely involve numerous peripheral hormones and central transmitters. In recent years, a prominent role of kisspeptins in the central pathways responsible for conveying metabolic information into the brain centers responsible for reproductive control, and specifically GnRH neurons, has been proposed on the basis of a wealth of expression and functional data. In this chapter, we will summarize such evidence, with special attention to the potential (direct and/or indirect) interaction of leptin and kisspeptin pathways. In addition, other potential metabolic modulators of kisspeptin signaling, as well as some of the putative molecular mechanisms for the metabolic regulation of Kiss1 will be briefly reviewed. Conflictive data, including those questioning an essential role of Kiss1 neurons in mediating leptin effects on the reproductive axis, will be also discussed. All in all, we aim to provide an integral and balanced view of the physiological relevance and potential mechanisms for the metabolic control of the kisspeptin system, as important pathway for the integral regulation of energy balance, puberty onset, and fertility.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético , Fertilidad , Kisspeptinas/metabolismo , Obesidad Mórbida/metabolismo , Pubertad , Reproducción , Transducción de Señal , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Leptina/metabolismo , Neuronas/metabolismo
4.
J Neurosci ; 30(23): 7783-92, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20534827

RESUMEN

The hypothalamic peptide, nesfatin-1, derived from the precursor NEFA/nucleobindin 2 (NUCB2), was recently identified as anorexigenic signal, acting in a leptin-independent manner. Yet its participation in the regulation of other biological functions gated by body energy status remains unexplored. We show herein that NUCB2/nesfatin-1 is involved in the control of female puberty. NUCB2/nesfatin mRNA and protein were detected at the hypothalamus of pubertal female rats, with prominent signals at lateral hypothalamus (LHA), paraventricular (PVN), and supraoptic (SON) nuclei. Hypothalamic NUCB2 expression raised along pubertal transition, with detectable elevations of its mRNA levels at LHA, PVN, and SON, and threefold increase of its total protein content between late-infantile and peripubertal periods. Conditions of negative energy balance, such as 48 h fasting or sustained subnutrition, decreased hypothalamic NUCB2 mRNA and/or protein levels in pubertal females. At this age, central administration of nesfatin-1 induced modest but significant elevations of circulating gonadotropins, whose magnitude was notably augmented in conditions of food deprivation. Continuous intracerebroventricular infusion of antisense morpholino oligonucleotides (as-MONs) against NUCB2 along pubertal maturation, which markedly reduced hypothalamic NUCB2 protein content, delayed vaginal opening and decreased ovarian weights and serum luteinizing hormone (LH) levels. In contrast, in adult female rats, intracerebroventricular injection of nesfatin did not stimulate LH or follicle-stimulating hormone secretion; neither did central as-MON infusion alter preovulatory gonadotropin surges, despite suppression of hypothalamic NUCB2. In sum, our data are the first to disclose the indispensable role of NUCB2/nesfatin-1 in the central networks driving puberty onset, a function that may contribute to its functional coupling to energy homeostasis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Neuropéptidos/farmacología , Maduración Sexual/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al ADN/genética , Femenino , Hormona Folículo Estimulante/sangre , Área Hipotalámica Lateral/metabolismo , Inyecciones Intraventriculares , Hormona Luteinizante/sangre , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/genética , Neuropéptidos/administración & dosificación , Neuropéptidos/metabolismo , Nucleobindinas , Oligorribonucleótidos Antisentido/administración & dosificación , Oligorribonucleótidos Antisentido/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero , Ratas , Ratas Wistar , Núcleo Supraóptico/metabolismo
5.
Cell Metab ; 32(6): 951-966.e8, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33080217

RESUMEN

Childhood obesity, especially in girls, is frequently bound to earlier puberty, which is linked to higher disease burden later in life. The mechanisms underlying this association remain elusive. Here we show that brain ceramides participate in the control of female puberty and contribute to its alteration in early-onset obesity in rats. Postnatal overweight caused earlier puberty and increased hypothalamic ceramide content, while pharmacological activation of ceramide synthesis mimicked the pubertal advancement caused by obesity, specifically in females. Conversely, central blockade of de novo ceramide synthesis delayed puberty and prevented the effects of the puberty-activating signal, kisspeptin. This phenomenon seemingly involves a circuit encompassing the paraventricular nucleus (PVN) and ovarian sympathetic innervation. Early-onset obesity enhanced PVN expression of SPTLC1, a key enzyme for ceramide synthesis, and advanced the maturation of the ovarian noradrenergic system. In turn, obesity-induced pubertal precocity was reversed by virogenetic suppression of SPTLC1 in the PVN. Our data unveil a pathway, linking kisspeptin, PVN ceramides, and sympathetic ovarian innervation, as key for obesity-induced pubertal precocity.


Asunto(s)
Ceramidas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Ovario/metabolismo , Obesidad Infantil , Pubertad Precoz , Animales , Femenino , Masculino , Obesidad Infantil/complicaciones , Obesidad Infantil/metabolismo , Pubertad Precoz/etiología , Pubertad Precoz/metabolismo , Ratas Wistar
6.
Environ Health Perspect ; 127(10): 107011, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31652106

RESUMEN

BACKGROUND: The timing of puberty is highly sensitive to environmental factors, including endocrine disruptors. Among them, bisphenol A (BPA) has been previously analyzed as potential modifier of puberty. Yet, disparate results have been reported, with BPA advancing, delaying, or being neutral in its effects on puberty onset. Likewise, mechanistic analyses addressing the central and peripheral actions/targets of BPA at puberty remain incomplete and conflictive. OBJECTIVE: We aimed to provide a comprehensive characterization of the impact of early BPA exposures, especially at low, real-life doses, on the postnatal development of hypothalamic Kiss1/NKB neurons, and its functional consequences on female pubertal maturation. METHODS: Pregnant CD1 female mice were orally administered BPA at 5, 10, or 40µg/kg body weight (BW)/d from gestational day 11 to postnatal day 8 (PND8). Vaginal opening, as an external marker of puberty onset, was monitored daily from PND19 to PND30 in the female offspring. Blood and brain samples were collected at PND12, 15, 18, 21, and 30 for measuring circulating levels of gonadotropins and analyzing the hypothalamic expression of Kiss1/kisspeptin and NKB. RESULTS: Perinatal exposure to BPA, in a range of doses largely below the no observed adverse effect level (NOAEL; 5mg/kg BW/d, according to the FDA), was associated with pubertal differences in the female progeny compared with those exposed to vehicle alone, with an earlier age of vaginal opening but consistently lower levels of circulating luteinizing hormone. Mice treated with BPA exhibited a persistent, but divergent, impairment of Kiss1 neuronal maturation, with more kisspeptin cells in the rostral (RP3V) hypothalamus but consistently fewer kisspeptin neurons in the arcuate nucleus (ARC). Detailed quantitative analysis of the ARC population, essential for pubertal development, revealed that mice treated with BPA had persistently lower Kiss1 expression during (pre)pubertal maturation, which was associated with lower Tac2 (encoding NKB) levels, even at low doses (5µg/kg BW/d), in the range of the tolerable daily intake (TDI), recently updated by the European Food Safety Authority. CONCLUSIONS: Our data attest to the consistent, but divergent, effects of gestational exposures to low concentrations of BPA, via the oral route, on phenotypic and neuroendocrine markers of puberty in female mice, with an unambiguous impact on the developmental maturation not only of Kiss1, but also of the NKB system, both essential regulators of puberty onset. https://doi.org/10.1289/EHP5570.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Contaminantes Ambientales/toxicidad , Kisspeptinas/metabolismo , Fenoles/toxicidad , Maduración Sexual/efectos de los fármacos , Animales , Disruptores Endocrinos , Femenino , Ratones , Neuronas/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Maduración Sexual/fisiología
7.
Endocrinology ; 159(2): 1005-1018, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29309558

RESUMEN

Obesity and its comorbidities are reaching epidemic proportions worldwide. Maternal obesity is known to predispose the offspring to metabolic disorders, independently of genetic inheritance. This intergenerational transmission has also been suggested for paternal obesity, with a potential negative impact on the metabolic and, eventually, reproductive health of the offspring, likely via epigenetic changes in spermatozoa. However, the neuroendocrine component of such phenomenon and whether paternal obesity sensitizes the offspring to the disturbances induced by high-fat diet (HFD) remain poorly defined. We report in this work the metabolic and reproductive impact of HFD in the offspring from obese fathers, with attention to potential sex differences and alterations of hypothalamic Kiss1 system. Lean and obese male rats were mated with lean virgin female rats; male and female offspring were fed HFD from weaning onward and analyzed at adulthood. The increases in body weight and leptin levels, but not glucose intolerance, induced by HFD were significantly augmented in the male, but not female, offspring from obese fathers. Paternal obesity caused a decrease in luteinizing hormone (LH) levels and exacerbated the drop in circulating testosterone and gene expression of its key biosynthetic enzymes caused by HFD in the male offspring. LH responses to central kisspeptin-10 administration were also suppressed in HFD males from obese fathers. In contrast, paternal obesity did not significantly alter gonadotropin levels in the female offspring fed HFD, although these females displayed reduced LH responses to kisspeptin-10. Our findings suggest that HFD-induced metabolic and reproductive disturbances are exacerbated by paternal obesity preferentially in males, whereas kisspeptin effects are affected in both sexes.


Asunto(s)
Padre , Kisspeptinas/fisiología , Obesidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Reproducción/fisiología , Animales , Femenino , Masculino , Obesidad/complicaciones , Embarazo , Ratas , Ratas Wistar , Salud Reproductiva , Caracteres Sexuales , Transducción de Señal/fisiología
8.
Nat Commun ; 6: 10195, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26671628

RESUMEN

In primates, puberty is unleashed by increased GnRH release from the hypothalamus following an interval of juvenile quiescence. GWAS implicates Zinc finger (ZNF) genes in timing human puberty. Here we show that hypothalamic expression of several ZNFs decreased in agonadal male monkeys in association with the pubertal reactivation of gonadotropin secretion. Expression of two of these ZNFs, GATAD1 and ZNF573, also decreases in peripubertal female monkeys. However, only GATAD1 abundance increases when gonadotropin secretion is suppressed during late infancy. Targeted delivery of GATAD1 or ZNF573 to the rat hypothalamus delays puberty by impairing the transition of a transcriptional network from an immature repressive epigenetic configuration to one of activation. GATAD1 represses transcription of two key puberty-related genes, KISS1 and TAC3, directly, and reduces the activating histone mark H3K4me2 at each promoter via recruitment of histone demethylase KDM1A. We conclude that GATAD1 epitomizes a subset of ZNFs involved in epigenetic repression of primate puberty.


Asunto(s)
Epigénesis Genética , Factores de Transcripción GATA/genética , Regulación del Desarrollo de la Expresión Génica , Hipotálamo/metabolismo , Pubertad/genética , ARN Mensajero/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Técnica del Anticuerpo Fluorescente , Hormona Folículo Estimulante/metabolismo , Factores de Transcripción GATA/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Histona Demetilasas/metabolismo , Hibridación Fluorescente in Situ , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Macaca mulatta , Masculino , Neuroquinina B/genética , Neuroquinina B/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc/genética
9.
Endocrinology ; 155(8): 3098-111, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24877631

RESUMEN

Neurotrophins (NTs), once believed to be neural-specific trophic factors, are now known to also provide developmental cues to non-neural cells. In the ovary, NTs contribute to both the formation and development of follicles. Here we show that oocyte-specific deletion of the Ntrk2 gene that encodes the NTRK2 receptor (NTRK2) for neurotrophin-4/5 and brain-derived neurotrophic factor (BDNF) results in post-pubertal oocyte death, loss of follicular organization, and early adulthood infertility. Oocytes lacking NTRK2 do not respond to gonadotropins with activation of phosphatidylinositol 3-kinase (PI3K)-AKT-mediated signaling. Before puberty, oocytes only express a truncated NTRK2 form (NTRK2.T1), but at puberty full-length (NTRK2.FL) receptors are rapidly induced by the preovulatory gonadotropin surge. A cell line expressing both NTRK2.T1 and the kisspeptin receptor (KISS1R) responds to BDNF stimulation with activation of Ntrk2 expression only if kisspeptin is present. This suggests that BDNF and kisspeptin that are produced by granulosa cells (GCs) of periovulatory follicles act in concert to mediate the effect of gonadotropins on Ntrk2 expression in oocytes. In keeping with this finding, the oocytes of NTRK2-intact mice fail to respond to gonadotropins with increased Ntrk2 expression in the absence of KISS1R. Our results demonstrate that the preovulatory gonadotropin surge promotes oocyte survival at the onset of reproductive cyclicity by inducing oocyte expression of NTRK2.FL receptors that set in motion an AKT-mediated survival pathway. They also suggest that gonadotropins activate NTRK2.FL expression via a dual communication pathway involving BDNF and kisspeptin produced in GCs and their respective receptors NTRK2.T1 and KISS1R expressed in oocytes.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Oocitos/metabolismo , Ovario/metabolismo , Insuficiencia Ovárica Primaria/etiología , Proteínas Tirosina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Gonadotropinas/fisiología , Infertilidad Femenina/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Kisspeptina-1
10.
Nat Neurosci ; 16(3): 281-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23354331

RESUMEN

The timing of puberty is controlled by many genes. The elements coordinating this process have not, however, been identified. Here we show that an epigenetic mechanism of transcriptional repression times the initiation of female puberty in rats. We identify silencers of the Polycomb group (PcG) as principal contributors to this mechanism and show that PcG proteins repress Kiss1, a puberty-activating gene. Hypothalamic expression of two key PcG genes, Eed and Cbx7, decreased and methylation of their promoters increased before puberty. Inhibiting DNA methylation blocked both events and resulted in pubertal failure. The pubertal increase in Kiss1 expression was accompanied by EED loss from the Kiss1 promoter and enrichment of histone H3 modifications associated with gene activation. Preventing the eviction of EED from the Kiss1 promoter disrupted pulsatile gonadotropin-releasing hormone release, delayed puberty and compromised fecundity. Our results identify epigenetic silencing as a mechanism underlying the neuroendocrine control of female puberty.


Asunto(s)
Epigénesis Genética , Hipotálamo/fisiología , Maduración Sexual/fisiología , Animales , Metilación de ADN , Estradiol/sangre , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Histonas/genética , Histonas/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Reproduction ; 133(6): 1223-32, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17636176

RESUMEN

Ghrelin, a gut peptide with key actions on food intake and GH secretion, has been recently recognized as potential regulator of reproductive function. Thus, in adult female rats, ghrelin has been proven to modulate GnRH/LH secretion, with predominant inhibitory effects in vivo. We analyze herein potential direct pituitary effects of ghrelin on basal and GnRH-stimulated gonadotropin secretion in prepubertal female rats, and its interplay with ovarian inputs, nitric oxide (NO), and hypothalamic differentiation. In the experimental setting, pituitaries from intact and ovariectomized prepubertal female rats were challenged with ghrelin in vitro and LH secretion was monitored. Our results demonstrate that 1) ghrelin consistently stimulated in vitro pituitary LH secretion under different experimental conditions; 2) the sensitivity to ghrelin, expressed either as the minimal effective dose or the amplitude of the LH response, was modulated by ovarian inputs; 3) the blockade of estrogen action significantly augmented the stimulatory effect of ghrelin; 4) the stimulatory effect of ghrelin on LH secretion required proper NO synthesis; and 5) the ability of ghrelin to elicit LH secretion in vitro was preserved after alteration (masculinization) of brain sexual differentiation. Overall, our present data reinforce the concept that ghrelin participates in the control of LH secretion, with potential stimulatory actions at the pituitary level that require the presence of NO and are modulated by ovarian signals.


Asunto(s)
Estrógenos/metabolismo , Hormona Luteinizante/metabolismo , Óxido Nítrico/metabolismo , Hormonas Peptídicas/farmacología , Hipófisis/metabolismo , Animales , Sinergismo Farmacológico , Femenino , Ghrelina , Hormona Liberadora de Gonadotropina/análisis , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Hormona Luteinizante/análisis , Ovariectomía , Hipófisis/efectos de los fármacos , Radioinmunoensayo , Ratas , Ratas Wistar , Estimulación Química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA