Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
PLoS Biol ; 17(11): e3000532, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31697675

RESUMEN

Mkrn3, the maternally imprinted gene encoding the makorin RING-finger protein-3, has recently emerged as putative pubertal repressor, as evidenced by central precocity caused by MKRN3 mutations in humans; yet, the molecular underpinnings of this key regulatory action remain largely unexplored. We report herein that the microRNA, miR-30, with three binding sites in a highly conserved region of its 3' UTR, operates as repressor of Mkrn3 to control pubertal onset. Hypothalamic miR-30b expression increased, while Mkrn3 mRNA and protein content decreased, during rat postnatal maturation. Neonatal estrogen exposure, causing pubertal alterations, enhanced hypothalamic Mkrn3 and suppressed miR-30b expression in female rats. Functional in vitro analyses demonstrated a strong repressive action of miR-30b on Mkrn3 3' UTR. Moreover, central infusion during the juvenile period of target site blockers, tailored to prevent miR-30 binding to Mkrn3 3' UTR, reversed the prepubertal down-regulation of hypothalamic Mkrn3 protein and delayed female puberty. Collectively, our data unveil a novel hypothalamic miRNA pathway, involving miR-30, with a prominent role in the control of puberty via Mkrn3 repression. These findings expand our current understanding of the molecular basis of puberty and its disease states.


Asunto(s)
Hipotálamo/metabolismo , MicroARNs/fisiología , Maduración Sexual/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Sitios de Unión , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , MicroARNs/metabolismo , Ratas , Análisis de Secuencia de ADN
3.
J Endocrinol ; 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31454785

RESUMEN

Human Prader-Willi syndrome (PWS) is characterised by impairments of multiple systems including the growth hormone (GH) axis and skeletal growth. To address our lack of knowledge of the influence of PWS on skeletal integrity in mice, we have characterised the endocrine and skeletal phenotype of the PWS-ICdel mouse model for "full" PWS and determined the impact of thermoneutrality. Tibial length, epiphyseal plate width and marrow adiposity were reduced by 6%, 18% and 79% in male PWS-ICdel mice, with osteoclast density being unaffected. Similar reductions in femoral length accompanied a 32% reduction in mid-diaphyseal cortical diameter. Distal femoral Tb.N was reduced by 62%, with individual trabeculae being less plate-like and the lattice being more fragmented (Tb.Pf increased by 63%). Cortical strength (Ultimate moment) was reduced by 26% as a result of reductions in calcified tissue strength and the geometric contribution. GH and prolactin contents in PWS-ICdel pituitaries were reduced in proportion to their smaller pituitary size, with circulating IGF-1 concentration reduced by 37-47%. Conversely, while pituitary LH content was halved, circulating gonadotropin concentrations were unaffected. Although longitudinal growth, marrow adiposity and femoral geometry were unaffected by thermoneutrality, strengthened calcified tissue reversed weakened cortex of PWS-ICdel femora. While underactivity of the GH-axis may be due to loss of Snord116 expression and impaired limb bone geometry and strength due to loss of Magel2 expression, comprehensive analysis of skeletal integrity in the single gene deletion models is required. Our data imply that thermoneutrality may ameliorate the elevated fracture risk associated with PWS.

4.
Metabolism ; 98: 84-94, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31226351

RESUMEN

BACKGROUND: Kisspeptins, encoded by Kiss1, have emerged as essential regulators of puberty and reproduction by primarily acting on GnRH neurons, via their canonical receptor, Gpr54. Mounting, as yet fragmentary, evidence strongly suggests that kisspeptin signaling may also participate in the control of key aspects of body energy and metabolic homeostasis. However, characterization of such metabolic dimension of kisspeptins remains uncomplete, without an unambiguous discrimination between the primary metabolic actions of kisspeptins vs. those derived from their ability to stimulate the secretion of gonadal hormones, which have distinct metabolic actions on their own. In this work, we aimed to tease apart primary vs. secondary effects of kisspeptins in the control of key aspects of metabolic homeostasis using genetic models of impaired kisspeptin signaling and/or gonadal hormone status. METHODS: Body weight (BW) gain and composition, food intake and key metabolic parameters, including glucose tolerance, were comparatively analyzed, in lean and obesogenic conditions, in mice lacking kisspeptin signaling due to global inactivation of Gpr54 (displaying profound hypogonadism; Gpr54-/-) vs. Gpr54 null mice with selective re-introduction of Gpr54 expression only in GnRH cells (Gpr54-/-Tg), where kisspeptin signaling elsewhere than in GnRH neurons is ablated but gonadal function is preserved. RESULTS: In male mice, global elimination of kisspeptin signaling resulted in decreased BW, feeding suppression and increased adiposity, without overt changes in glucose tolerance, whereas Gpr54-/- female mice displayed enhanced BW gain at adulthood, increased adiposity and perturbed glucose tolerance, despite reduced food intake. Gpr54-/-Tg rescued mice showed altered postnatal BW gain in males and mildly perturbed glucose tolerance in females, with intermediate phenotypes between control and global KO animals. Yet, body composition and leptin levels were similar to controls in gonadal-rescued mice. Exposure to obesogenic insults, such as high fat diet (HFD), resulted in exaggerated BW gain and adiposity in global Gpr54-/- mice of both sexes, and worsening of glucose tolerance, especially in females. Yet, while rescued Gpr54-/-Tg males displayed intermediate BW gain and feeding profiles and impaired glucose tolerance, rescued Gpr54-/-Tg females behaved as controls, except for a modest deterioration of glucose tolerance after ovariectomy. CONCLUSION: Our data support a global role of kisspeptin signaling in the control of body weight and metabolic homeostasis, with a dominant contribution of gonadal hormone-dependent actions. However, our results document also discernible primary effects of kisspeptin signaling in the regulation of body weight gain, feeding and responses to obesogenic insults, which occur in a sexually-dimorphic manner. SUMMARY OF TRANSLATIONAL RELEVANCE: Kisspeptins, master regulators of reproduction, may also participate in the control of key aspects of body energy and metabolic homeostasis; yet, the nature of such metabolic actions remains debatable, due in part to the fact that kisspeptins modulate gonadal hormones, which have metabolic actions on their own. By comparing the metabolic profiles of two mouse models with genetic inactivation of kisspeptin signaling but different gonadal status (hypogonadal vs. preserved gonadal function), we provide herein a systematic dissection of gonadal-dependent vs. -independent metabolic actions of kisspeptins. Our data support a global role of kisspeptin signaling in the control of body weight and metabolic homeostasis, with a dominant contribution of gonadal hormone-dependent actions. However, our results document also discernible primary effects of kisspeptin signaling in the regulation of body weight gain, feeding and responses to obesogenic insults, which occur in a sexually-dimorphic manner. These data pave the way for future analyses addressing the eventual contribution of altered kisspeptin signaling in the development of metabolic alterations, especially in conditions linked to reproductive dysfunction.


Asunto(s)
Peso Corporal/fisiología , Hormonas Gonadales/fisiología , Homeostasis/fisiología , Kisspeptinas/fisiología , Transducción de Señal/fisiología , Animales , Dieta , Ingestión de Alimentos , Femenino , Intolerancia a la Glucosa/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Ovariectomía , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Aumento de Peso/genética
5.
Proc Natl Acad Sci U S A ; 115(45): E10758-E10767, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348767

RESUMEN

Conditions of metabolic distress, from malnutrition to obesity, impact, via as yet ill-defined mechanisms, the timing of puberty, whose alterations can hamper later cardiometabolic health and even life expectancy. AMP-activated protein kinase (AMPK), the master cellular energy sensor activated in conditions of energy insufficiency, has a major central role in whole-body energy homeostasis. However, whether brain AMPK metabolically modulates puberty onset remains unknown. We report here that central AMPK interplays with the puberty-activating gene, Kiss1, to control puberty onset. Pubertal subnutrition, which delayed puberty, enhanced hypothalamic pAMPK levels, while activation of brain AMPK in immature female rats substantially deferred puberty. Virogenetic overexpression of a constitutively active form of AMPK, selectively in the hypothalamic arcuate nucleus (ARC), which holds a key population of Kiss1 neurons, partially delayed puberty onset and reduced luteinizing hormone levels. ARC Kiss1 neurons were found to express pAMPK, and activation of AMPK reduced ARC Kiss1 expression. The physiological relevance of this pathway was attested by conditional ablation of the AMPKα1 subunit in Kiss1 cells, which largely prevented the delay in puberty onset caused by chronic subnutrition. Our data demonstrate that hypothalamic AMPK signaling plays a key role in the metabolic control of puberty, acting via a repressive modulation of ARC Kiss1 neurons in conditions of negative energy balance.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Desnutrición/metabolismo , Neuronas/metabolismo , Maduración Sexual/genética , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Animales Modificados Genéticamente , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Restricción Calórica/efectos adversos , Estradiol/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Kisspeptinas/genética , Hormona Luteinizante/sangre , Desnutrición/genética , Desnutrición/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Ribonucleótidos/farmacología , Transducción de Señal , Factores de Tiempo
6.
Sci Rep ; 7: 46381, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28401948

RESUMEN

Puberty is a key developmental event whose primary regulatory mechanisms remain poorly understood. Precise dating of puberty is crucial for experimental (preclinical) studies on its complex neuroendocrine controlling networks. In female laboratory rodents, external signs of puberty, such as vaginal opening (VO) and epithelial cell cornification (i.e., first vaginal estrus, FE), are indirectly related to the maturational state of the ovary and first ovulation, which is the unequivocal marker of puberty. Whereas in rats, VO and FE are almost simultaneous with the first ovulation, these events are not so closely associated in mice. Moreover, external signs of puberty can be uncoupled with first ovulation in both species under certain experimental conditions. We propose herein the Pubertal Ovarian Maturation Score (Pub-score), as novel, reliable method to assess peripubertal ovarian maturation in rats and mice. This method is founded on histological evaluation of pre-pubertal ovarian maturation, based on antral follicle development, and the precise timing of first ovulation, by retrospective dating of maturational and regressive changes in corpora lutea. This approach allows exact timing of puberty within a time-window of at least two weeks after VO in both species, thus facilitating the identification and precise dating of advanced or delayed puberty under various experimental conditions.


Asunto(s)
Estro/fisiología , Ovulación/fisiología , Maduración Sexual/fisiología , Vagina/fisiología , Animales , Animales de Laboratorio , Femenino , Ratones , Ratas , Factores de Tiempo
7.
Expert Opin Ther Targets ; 20(10): 1181-93, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27409160

RESUMEN

INTRODUCTION: The onset of puberty in females is highly sensitive to the nutritional status and the amount of energy reserves of the organism. This metabolic information is sensed and transmitted to hypothalamic GnRH neurons, considered to be ultimately responsible for triggering puberty through the coordinated action of different peripheral hormones, central neurotransmitters, and molecular mediators. AREAS COVERED: This article will review and discuss (i) the relevant actions of the adipose hormone leptin, as a stimulatory/permissive signal, and the gut hormone ghrelin, as an inhibitory factor, in the metabolic control of female puberty; (ii) the crucial role of the hypothalamic kisspeptin neurons, recently emerged as essential gatekeepers of puberty, in transmitting this metabolic information to GnRH neurons; and (iii) the potential involvement of key cellular energy sensors, such as mTOR, as molecular mediators in this setting. EXPERT OPINION: The thorough characterization of the physiological roles of the above elements in the metabolic control of female puberty, along with the discovery of novel factors, pathways, and mechanisms involved, will promote our understanding of the complex networks connecting metabolism and puberty and, ultimately, will aid in the design of target-specific treatments for female pubertal disorders linked to conditions of metabolic stress.


Asunto(s)
Hipotálamo/metabolismo , Estado Nutricional/fisiología , Pubertad/metabolismo , Animales , Femenino , Ghrelina/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Leptina/metabolismo , Neuronas/metabolismo , Maduración Sexual/fisiología
8.
Endocr Dev ; 29: 87-121, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26680574

RESUMEN

Puberty is a fascinating developmental transition that gates the attainment of reproductive capacity and culminates the somatic and sexual maturation of the organism. Rather than a circumscribed phenomenon, puberty is the endpoint of a long-lasting developmental continuum, which initiates in utero. Besides important genetic determinants, the tempo of puberty is influenced by numerous endogenous and exogenous factors that, acting at different levels of the developing hypothalamic-pituitary-gonadal (HPG) axis along the maturational continuum indicated above, can influence puberty onset. Among the different modifiers of puberty, in this chapter we will focus our attention on two major groups of signals, sex steroids and nutritional cues, and how these interplay mostly with the central elements of the HPG axis, and especially with gonadotropin-releasing hormone neurons and their key upstream afferents, Kiss1 neurons, to influence the timing of puberty. Special emphasis will be given to summarize information emerging from relevant preclinical (mostly rodent) animal models, and how this information might be relevant in terms of translational medicine, as it may help for a better understanding and eventually management of pubertal disorders of escalating prevalence worldwide.


Asunto(s)
Pubertad Tardía/genética , Pubertad Precoz/genética , Pubertad/genética , Maduración Sexual/genética , Animales , Modelos Animales de Enfermedad , Femenino , Gónadas/crecimiento & desarrollo , Humanos , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Masculino
9.
Endocrinology ; 156(8): 2984-98, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25984764

RESUMEN

The reproductive impact of persistent energy excess in the female remains incompletely defined, yet the escalating prevalence of obesity calls for better understanding of this phenomenon. Also along this line, the influence of ovarian hormones on the pathophysiology of obesity and its comorbidities merits further investigation. We study here the metabolic and gonadotropic impact of sequential obesogenic insults, namely postnatal overnutrition [by rearing in small litters (SL)] and high-fat diet (HFD) after weaning, in gonadal-intact and ovariectomized (OVX) female rats. In young (4 mo) females, SL or HFD similarly increased body weight, yet only a HFD evoked additional metabolic perturbations, some of which were worsened by precedent SL. In addition, HFD concomitantly decreased LH and estradiol levels and, when combined with SL, suppressed Kiss1 expression in the hypothalamic arcuate nucleus in 4-month females, whereas HFD up to 10-month also reduced LH responses to kisspeptin-10. OVX caused rapid deterioration of the metabolic profile, with overweight, increased energy intake, and deregulation of leptin and glucose/insulin levels, effects whose magnitude was similar to, if not higher than, HFD. Summation of previous obesogenic insults maximally increased body weight, basal leptin, insulin and glucose levels, and glucose intolerance. Yet OVX obliterated the inhibitory effects of overweight/HFD on gonadotropin levels and arcuate nucleus Kiss1 expression. Our study documents the deleterious consequences of sequential obesogenic insults on the female gonadotropin axis, which involve central impairment of the Kiss1 system. In addition, our work delineates the dramatic impact of the loss of ovarian secretions, as the menopausal model, on the metabolic profile of female rats, especially when combined with preceding obesogenic challenges.


Asunto(s)
Dieta Alta en Grasa , Gonadotrofos/fisiología , Ovario/metabolismo , Hipernutrición/metabolismo , Hipernutrición/fisiopatología , Envejecimiento/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Gonadotrofos/metabolismo , Fenómenos Fisiológicos de la Nutrición , Obesidad/etiología , Obesidad/metabolismo , Obesidad/fisiopatología , Ovario/fisiopatología , Ratas , Ratas Wistar , Reproducción/fisiología
10.
Endocrinology ; 155(10): 3945-55, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25051440

RESUMEN

Kisspeptin, neurokinin B (NKB) and dynorphin A are coexpressed in a population of neurons in the arcuate nucleus (ARC), termed KNDy neurons, which were recently recognized as important elements for the generation of GnRH pulses. However, the topographic distribution of these peptides and their regulated expression by sex steroids are still not well understood. In this study, detailed examination of NKB and kisspeptin immunoreactivity in the rat ARC was carried out, including comparison between sexes, with and without sex steroid replacement. Neurons expressing kisspeptin and NKB were more prominent in the caudal ARC of females, whereas neurons expressing NKB, but not kisspeptin, were the most abundant in the male. Sex steroid manipulation revealed differential regulation of kisspeptin and NKB; although kisspeptin immunoreactive (ir) cells increased in response to gonadectomy, NKB remained unchanged. Furthermore, the number of NKB-ir cells increased upon sex steroid replacement compared with gonadectomy, whereas kisspeptin did not, suggesting that sex steroids differently regulate these peptides. In addition, only in females did the density of kisspeptin- and NKB-ir fibers in the ARC increase upon sex steroid replacement in relation to sham and ovariectomy, respectively, suggesting sex-specific regulation of release. In conclusion, our observations reveal sex differences in the number of kisspeptin- and NKB-ir cells, which are more prominent in the caudal ARC. The divergent regulation of kisspeptin and NKB peptide contents in the ARC as a function of sex and steroid milieu enlarge our understanding on how these neuropeptides are posttranscriptionally regulated in KNDy neurons.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormonas Esteroides Gonadales/fisiología , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Castración , Recuento de Células , Dinorfinas/metabolismo , Femenino , Hormonas Esteroides Gonadales/farmacología , Kisspeptinas/genética , Masculino , Neuroquinina B/genética , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Wistar , Caracteres Sexuales
11.
J Clin Endocrinol Metab ; 99(10): E2067-75, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25033069

RESUMEN

CONTEXT: Gordon Holmes syndrome (GHS) is characterized by cerebellar ataxia/atrophy and normosmic hypogonadotropic hypogonadism (nHH). The underlying pathophysiology of this combined neurodegeneration and nHH remains unknown. OBJECTIVE: We aimed to provide insight into the disease mechanism in GHS. METHODS: We studied a cohort of 6 multiplex families with GHS through autozygosity mapping and whole-exome sequencing. RESULTS: We identified 6 patients from 3 independent families carrying loss-of-function mutations in PNPLA6, which encodes neuropathy target esterase (NTE), a lysophospholipase that maintains intracellular phospholipid homeostasis by converting lysophosphatidylcholine to glycerophosphocholine. Wild-type PNPLA6, but not PNPLA6 bearing these mutations, rescued a well-established Drosophila neurodegenerative phenotype caused by the absence of sws, the fly ortholog of mammalian PNPLA6. Inhibition of NTE activity in the LßT2 gonadotrope cell line diminished LH response to GnRH by reducing GnRH-stimulated LH exocytosis, without affecting GnRH receptor signaling or LHß synthesis. CONCLUSION: These results suggest that NTE-dependent alteration of phospholipid homeostasis in GHS causes both neurodegeneration and impaired LH release from pituitary gonadotropes, leading to nHH.


Asunto(s)
Ataxia Cerebelosa/genética , Hormona Liberadora de Gonadotropina/deficiencia , Hipogonadismo/genética , Degeneración Nerviosa/genética , Fosfolipasas/genética , Pubertad Tardía/genética , Adolescente , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Ataxia Cerebelosa/metabolismo , Salud de la Familia , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Homeostasis/genética , Humanos , Hipogonadismo/metabolismo , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/metabolismo , Linaje , Fosfolipasas/metabolismo , Fosfolípidos/metabolismo , Pubertad Tardía/metabolismo
12.
Endocrinology ; 155(8): 3088-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24885574

RESUMEN

Premature ovarian failure (POF) affects 1% of women in reproductive age, but its etiology remains uncertain. Whereas kisspeptins, the products of Kiss1 that act via Kiss1r (aka, Gpr54), are known to operate at the hypothalamus to control GnRH/gonadotropin secretion, additional actions at other reproductive organs, including the ovary, have been proposed. Yet, their physiological relevance is still unclear. We present here a series of studies in Kiss1r haplo-insufficient and null mice suggesting a direct role of kisspeptin signaling in the ovary, the defect of which precipitates a state of primary POF. Kiss1r hypomorph mice displayed a premature decline in ovulatory rate, followed by progressive loss of antral follicles, oocyte loss, and a reduction in all categories of preantral follicles. These alterations were accompanied by reduced fertility. Because of this precocious ovarian ageing, mice more than 48 weeks of age showed atrophic ovaries, lacking growing follicles and corpora lutea. This phenomenon was associated with a drop in ovarian Kiss1r mRNA expression, but took place in the absence of a decrease in circulating gonadotropins. In fact, FSH levels increased in aged hypomorph animals, reflecting loss of follicular function. In turn, Kiss1r-null mice, which do not spontaneously ovulate and have arrested follicular development, failed to show normal ovulatory responses to standard gonadotropin priming and required GnRH prestimulation during 1 week in order to display gonadotropin-induced ovulation. Yet, the magnitude of such ovulatory responses was approximately half of that seen in control immature wild-type animals. Altogether, our data are the first to demonstrate that Kiss1r haplo-insufficiency induces a state of POF, which is not attributable to defective gonadotropin secretion. We also show that the failure of follicular development and ovulation linked to the absence of Kiss1r cannot be fully rescued by (even extended) gonadotropin replacement. These findings suggest a direct ovarian role of kisspeptin signaling, the perturbation of which may contribute to the pathogenesis of POF.


Asunto(s)
Kisspeptinas/metabolismo , Ovario/fisiología , Ovulación , Insuficiencia Ovárica Primaria/etiología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Femenino , Gonadotropinas/metabolismo , Hipogonadismo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptores de Kisspeptina-1
13.
Endocrinology ; 155(3): 1067-79, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424048

RESUMEN

Reproduction is sensitive to insufficient body energy reserves, especially in females. Metabolic regulation of the male reproductive axis is less obvious, and the impact of conditions of persistent energy excess has received moderate attention. Yet, the escalating prevalence of obesity and the clinical evidence of its deleterious effects on male fertility have raised considerable concerns. We report here phenotypic and mechanistic studies of the reproductive impact of postnatal nutritional manipulations (mainly overnutrition) coupled to a high-fat diet (HFD) after weaning. Metabolic and hormonal analyses in young (4 months old) and middle-aged (10 months old) animals revealed that HFD caused profound metabolic perturbations, including glucose intolerance, which were worsened by precedent postnatal overfeeding; these were detectable already in young males but aggravated in 10-month-old rats. Impairment of reproductive parameters took place progressively, and HFD alone was sufficient to explain most of these alterations, regardless of postnatal under- or overnutrition. In young males, testosterone (T) levels and steroidogenic enzyme expression were suppressed by HFD, without compensatory increases of LH levels, which were in fact partially inhibited in heavier males. In addition, obese males displayed suppressed hypothalamic Kiss1 expression despite low T, and HFD inhibited LH responses to kisspeptin. Overweight anticipated some of the neuroendocrine effects of aging, such as the suppression of hypothalamic Kiss1 expression and the decline in serum T and LH levels. Nonetheless, HFD per se caused a detectable worsening of key reproductive indices in middle-aged males, such as basal LH and FSH levels as well as LH responses to kisspeptin. Our study demonstrates that nutritional stress, especially HFD, has a profound deleterious impact on metabolic and gonadotropic function as well as on the Kiss1 system and precipitates neuroendocrine reproductive senescence in the male.


Asunto(s)
Dieta Alta en Grasa , Hipogonadismo/metabolismo , Hipogonadismo/patología , Kisspeptinas/metabolismo , Sistemas Neurosecretores/fisiología , Obesidad/patología , Animales , Peso Corporal , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hipogonadismo/etiología , Hipotálamo/metabolismo , Hibridación in Situ , Hormona Luteinizante/sangre , Masculino , Obesidad/complicaciones , Fenotipo , Ratas , Ratas Wistar , Reproducción , Factores Sexuales , Testosterona/metabolismo , Factores de Tiempo
14.
Am J Physiol Endocrinol Metab ; 303(10): E1252-63, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23011064

RESUMEN

Kisspeptins (Kp), products of the Kiss1 gene, have emerged as essential elements in the control of GnRH neurons and gonadotropic secretion. However, despite considerable progress in the field, to date limited attention has been paid to elucidate the potential interactions of Kp with other neurotransmitters known to centrally regulate the gonadotropic axis. We characterize herein the impact of manipulations of key aminoacidergic (glutamate and GABA), peptidergic (NKB, Dyn, and MCH), and gaseous [nitric oxide (NO)] neurotransmission on gonadotropin responses to Kp-10 in male rats. Blockade of ionotropic glutamate receptors (of the NMDA and non-NMDA type) variably decreased LH responses to Kp-10, whereas activation of both ionotropic and metabotropic receptors, which enhanced LH and FSH release per se, failed to further increase gonadotropin responses to Kp-10. In fact, coactivation of metabotropic receptors attenuated LH and FSH responses to Kp-10. Selective activation of GABA(A) receptors decreased Kp-induced gonadotropin secretion, whereas their blockade elicited robust LH and FSH bursts and protracted responses to Kp-10 when combined with GABA(B) receptor inhibition. Blockade of Dyn signaling (at κ-opioid receptors) enhanced LH responses to Kp-10, whereas activation of Dyn and NKB signaling modestly reduced Kp-induced LH and FSH release. Finally, MCH decreased basal LH secretion and modestly reduced FSH responses to Kp-10, whereas LH responses to Kp-10 were protracted after inhibition of NO synthesis. In summary, we present herein evidence for the putative roles of glutamate, GABA, Dyn, NKB, MCH, and NO in modulating gonadotropic responses to Kp in male rats. Our pharmacological data will help to characterize the central interactions and putative hierarchy of key neuroendocrine pathways involved in the control of the gonadotropic axis.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Kisspeptinas/farmacología , Hormona Luteinizante/metabolismo , Transmisión Sináptica/efectos de los fármacos , Animales , Dineínas/antagonistas & inhibidores , Dineínas/metabolismo , Hormona Folículo Estimulante/sangre , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/agonistas , Hormonas Hipotalámicas/antagonistas & inhibidores , Hormonas Hipotalámicas/metabolismo , Hormona Luteinizante/sangre , Masculino , Melaninas/agonistas , Melaninas/antagonistas & inhibidores , Melaninas/metabolismo , Neuroquinina B/agonistas , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Hormonas Hipofisarias/agonistas , Hormonas Hipofisarias/antagonistas & inhibidores , Hormonas Hipofisarias/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
15.
J Neurosci ; 32(7): 2388-97, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22396413

RESUMEN

Human genetic studies have revealed that neurokinin B (NKB) and its receptor, neurokinin-3 receptor (NK3R), are essential elements for normal reproduction; however, the precise role of NKB-NK3R signaling in the initiation of puberty remains unknown. We investigated here the regulation of Tac2 and Tacr3 mRNAs (encoding NKB and NK3R, respectively) in female rats and demonstrated that their hypothalamic expression is increased along postnatal maturation. At puberty, both genes were widely expressed throughout the brain, including the lateral hypothalamic area and the arcuate nucleus (ARC)/medial basal hypothalamus, where the expression of Tacr3 increased across pubertal transition. We showed that central administration of senktide (NK3R agonist) induced luteinizing hormone (LH) secretion in prepubertal and peripubertal females. Conversely, chronic infusion of an NK3R antagonist during puberty moderately delayed the timing of vaginal opening (VO) and tended to decrease LH levels. The expression of NKB and its receptor was sensitive to changes in metabolic status during puberty, as reflected by a reduction in Tacr3 (and, to a lesser extent, Tac2) expression in the ARC after a 48 h fast. Yet, acute LH responses to senktide in pubertal females were preserved, if not augmented, under fasting conditions, suggesting sensitization of the NKB-NK3R-gonadotropin-releasing hormone signaling pathway under metabolic distress. Moreover, repeated administration of senktide to female rats with pubertal arrest due to chronic undernutrition rescued VO (in ∼50% of animals) and potently elicited LH release. Altogether, our observations suggest that NKB-NK3R signaling plays a role in pubertal maturation and that its alterations may contribute to pubertal disorders linked to metabolic stress and negative energy balance.


Asunto(s)
Metaboloma/fisiología , Neuroquinina B/fisiología , Maduración Sexual/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/fisiología , Femenino , Neuroquinina B/metabolismo , Ratas , Ratas Wistar , Receptores de Neuroquinina-3/metabolismo , Receptores de Neuroquinina-3/fisiología
16.
Mol Cell Endocrinol ; 351(2): 184-98, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22209758

RESUMEN

Mammalian puberty is initiated by an increased pulsatile release of gonadotropin-releasing hormone (GnRH) from specialized neurons located in the hypothalamus. GnRH secretion is controlled by neuronal and glial networks, whose activity appears to be coordinated via transcriptional regulation. One of the transcription factors involved in this process is thought to be the recently described gene Enhanced at Puberty 1 (EAP1), which encodes a protein with dual transcriptional activity. In this study we used gene reporter and chromatin immunoprecipitation (ChIP) assays to examine the hypothesis that EAP1 expression is controlled by transcriptional regulators earlier postulated to serve as central nodes of a gene network involved in the neuroendocrine control of puberty. These regulators include Thyroid Transcription Factor 1 (TTF1), Yin Yang 1 (YY1), and CUX1, in addition to EAP1 itself. While TTF1 has been shown to facilitate the advent of puberty, YY1 (a zinc finger protein component of the Polycomb silencing complex) may play a repressive role. The precise role of CUX1 in this context is not known, but like EAP1, CUX1 can either activate or repress gene transcription. We observed that DNA segments of two different lengths (998 and 2744bp) derived from the 5'-flanking region of the human EAP1 gene display similar transcriptional activity. TTF1 stimulates transcription from both DNA segments with equal potency, whereas YY1, CUX1, and EAP1 itself, behave as transcriptional repressors. All four proteins are recruited in vivo to the EAP1 5'-flanking region. These observations suggest that EAP1 gene expression is under dual transcriptional regulation imposed by a trans-activator (TTF1) and two repressors (YY1 and CUX1) previously postulated to be upstream components of a puberty-controlling gene network. In addition, EAP1 itself appears to control its own expression via a negative auto-feedback loop mechanism. Further studies are needed to determine if the occupancy of the EAP1 promoter by these regulatory factors changes at the time of puberty.


Asunto(s)
Redes Reguladoras de Genes , Genes Reguladores/genética , Genes Supresores de Tumor , Proteínas de Neoplasias/genética , Animales , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Proteínas de Neoplasias/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Pubertad/genética , Ratas , Ratas Sprague-Dawley , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Securina , Factores de Transcripción , Transcripción Genética , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
17.
Endocrinology ; 152(9): 3396-408, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21712362

RESUMEN

Kiss1 neurons have recently emerged as a putative conduit for the metabolic gating of reproduction, with leptin being a regulator of hypothalamic Kiss1 expression. Early perturbations of the nutritional status are known to predispose to different metabolic disorders later in life and to alter the timing of puberty; however, the potential underlying mechanisms remain poorly defined. Here we report how changes in the pattern of postnatal feeding affect the onset of puberty and evaluate key hormonal and neuropeptide [Kiss1/kisspeptin (Kp)] alterations linked to these early nutritional manipulations. Female rats were raised in litters of different sizes: small (four pups per dam: overfeeding), normal (12 pups per dam), and large litters (20 pups per litter: underfeeding). Postnatal overfeeding resulted in persistently increased body weight and earlier age of vaginal opening, as an external sign of puberty, together with higher levels of leptin and hypothalamic Kiss1 mRNA. Conversely, postnatal underfeeding caused a persistent reduction in body weight, lower ovarian and uterus weights, and delayed vaginal opening, changes that were paralleled by a decrease in leptin and Kiss1 mRNA levels. Kisspeptin-52 immunoreactivity (Kp-IR) in the hypothalamus displayed similar patterns, with lower numbers of Kp-IR neurons in the arcuate nucleus of postnatally underfed animals, and a trend for increased Kp-positive fibers in the periventricular area of early overfed rats. Yet, gonadotropin responses to Kp at puberty were similar in all groups, except for enhanced responsiveness to low doses of Kp-10 in postnatally underfed rats. In conclusion, our data document that the timing of puberty is sensitive to both overfeeding and subnutrition during early (postnatal) periods and suggest that alterations in hypothalamic expression of Kiss1/kisspeptin may underlie at least part of such programming phenomenon.


Asunto(s)
Hipotálamo/metabolismo , Conducta Materna/fisiología , Proteínas/metabolismo , Maduración Sexual/fisiología , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Femenino , Kisspeptinas , Leptina/sangre , Hormona Luteinizante/sangre , Neuronas/metabolismo , Ratas , Ratas Wistar
18.
Am J Physiol Endocrinol Metab ; 300(1): E202-10, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21045176

RESUMEN

Neurokinin B (NKB) and its cognate receptor neurokinin 3 (NK3R) play a critical role in reproduction. NKB and NK3R are coexpressed with dynorphin (Dyn) and kisspeptin (Kiss1) genes in neurons of the arcuate nucleus (Arc). However, the mechanisms of action of NKB as a cotransmitter with kisspeptin and dynorphin remain poorly understood. We explored the role of NKB in the control of LH secretion in the female rat as follows. 1) We examined the effect of an NKB agonist (senktide, 600 pmol, administered into the lateral cerebral ventricle) on luteinizing hormone (LH) secretion. In the presence of physiological levels of estradiol (E(2)), senktide induced a profound increase in serum levels of LH and a 10-fold increase in the number of Kiss1 neurons expressing c-fos in the Arc (P < 0.01 for both). 2) We mapped the distribution of NKB and NK3R mRNAs in the central forebrain and found that both are widely expressed, with intense expression in several hypothalamic nuclei that control reproduction, including the Arc. 3) We studied the effect of E(2) on the expression of NKB and NK3R mRNAs in the Arc and found that E(2) inhibits the expression of both genes (P < 0.01) and that the expression of NKB and NK3R reaches its nadir on the afternoon of proestrus (when circulating levels of E(2) are high). These observations suggest that NKB/NK3R signaling in Kiss1/NKB/Dyn-producing neurons in the Arc has a pivotal role in the control of gonadotropin-releasing hormone (GnRH)/LH secretion and its regulation by E(2)-dependent negative feedback in the rat.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Proteínas/metabolismo , Receptores de Neuroquinina-3/metabolismo , Transducción de Señal , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Estradiol/metabolismo , Ciclo Estral/metabolismo , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica , Kisspeptinas , Hormona Luteinizante/sangre , Neuroquinina B/agonistas , Neuroquinina B/genética , Neuronas/efectos de los fármacos , Especificidad de Órganos , Fragmentos de Péptidos/farmacología , Prosencéfalo/citología , Prosencéfalo/metabolismo , Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/genética , Transducción de Señal/efectos de los fármacos , Sustancia P/análogos & derivados , Sustancia P/farmacología
19.
Brain Res ; 1364: 129-38, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20800054

RESUMEN

Body energy reserves and metabolic state are relevant modifiers of puberty onset and fertility; forms of metabolic stress ranging from persistent energy insufficiency to morbid obesity are frequently linked to reproductive disorders. The mechanisms for such a close connection between energy balance and reproduction have been the subject of considerable attention; however, our understanding of the neurobiological basis for this phenomenon is still incomplete. In mid 1990s, the adipose-hormone, leptin, was proven as an essential signal for transmitting metabolic information onto the centers governing puberty and reproduction; yet, the ultimate mode of action of leptin on GnRH neurons has remained contentious for years. More recently, kisspeptins, a family of neuropeptides encoded by the Kiss1 gene, have emerged as conduits for the metabolic regulation of reproduction and putative effectors of leptin actions on GnRH neurons. This review recapitulates the experimental evidence obtained to date, mostly in laboratory rodents, supporting the function of kisspeptins in bridging energy balance and reproduction, with special emphasis on recent developments in this field, such as the recognition of mTOR (mammalian target of rapamycin) and Crtc1 (Creb1-regulated transcription coactivator-1) as putative mediators for leptin regulation of Kiss1 expression, as well as the identification of other potential metabolic modulators of kisspeptin signaling, such as ghrelin, neuropeptide Y (NPY) and melanin-concentrating hormone (MCH).


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis/fisiología , Reproducción/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Fertilidad/fisiología , Humanos , Kisspeptinas , Leptina/fisiología , Pubertad/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética
20.
Mol Cell Endocrinol ; 324(1-2): 87-94, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20026241

RESUMEN

Puberty, as the end-point of a complex series of maturational events affecting the components of the hypothalamic-pituitary-gonadal (HPG) axis, is gated by the state of body energy reserves and sensitive to different metabolic cues; conditions of severe metabolic stress and energy unbalance (from anorexia to morbid obesity) being commonly linked to perturbation of the onset of puberty. In the last two decades, the neuroendocrine mechanisms responsible for the tight coupling between energy homeostasis and puberty onset have begun to be deciphered. These seemingly involve a plethora of metabolic hormones and neuropeptides, which impinge and integrate (mostly) at the hypothalamic centers governing reproduction. Yet, characterization of the mechanisms of action of such regulators (and even their nature and physiological relevance) still remains incomplete. In this review, we will summarize some recent developments in our knowledge of the effects and mechanisms of action of two key metabolic hormones, leptin and ghrelin, in the control of puberty onset. In addition, the roles of the hypothalamic Kiss1 system in the metabolic gating of puberty will be reviewed, with special attention to its regulation by leptin and the recent identification of the putative roles of Crtc1 and mTOR signaling as molecular conduits for the metabolic control of Kiss1 expression. Elucidation of these novel players and regulatory mechanisms will help for a better understanding of the determinants of the timing of puberty, and its eventual alterations in adverse metabolic conditions.


Asunto(s)
Pubertad/metabolismo , Maduración Sexual/fisiología , Animales , Ghrelina/metabolismo , Humanos , Leptina/metabolismo , Obesidad/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...