Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 573
Filtrar
1.
Brain Res ; 1727: 146563, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31765630

RESUMEN

Even though several of RFamide peptides have been shown to modify memory and learning processes in different species, almost nothing is known regarding cognitive effects of recently discovered neuropeptide QRFP. Considering multiple physiological functions of QRFP, localization of QRFP-synthesizing neurons in the hypothalamus and its' widely spread binding sites within the CNS, the present study was designed to investigate the possible role of QRFP in the consolidation of spatial memory. As target area for microinjection, the medial hypothalamic area, including dorsomedial (DMN) and ventromedial (VMN) nuclei, has been chosen. At first, the effects of two doses (200 ng and 400 ng) of QRFP were investigated in Morris water maze. After that receptor antagonist BIBP3226 (equimolar amount to the effective dose of neuropeptide) was applied to elucidate whether it can prevent effects of QRFP. To reveal possible changes in anxiety level, animals were tested in Elevated plus maze. The higher dose of QRFP (400 ng) improved short-term memory consolidation in Morris water maze. Pretreatment with antagonist BIBP3226 abolished cognitive effects of QRFP. The neuropeptide did not affect anxiety level of rats. This study provides unique evidence regarding the role of QRFP in the consolidation of memory and gives the basis for further investigations of neuropeptide's cognitive effects.


Asunto(s)
Hipotálamo Medio/fisiología , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/fisiología , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Hipotálamo Medio/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratas Wistar
2.
J Neuroendocrinol ; 31(12): e12810, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31715027

RESUMEN

The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.


Asunto(s)
Dinorfinas/metabolismo , Hipotálamo Medio/metabolismo , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Microinyecciones , Neuroquinina B/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Ratas , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptores de Neuroquinina-3/biosíntesis , Receptores Opioides/biosíntesis , Maduración Sexual , Sustancia P/análogos & derivados , Sustancia P/farmacología , Regulación hacia Arriba
3.
Brain Res ; 1725: 146468, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31541642

RESUMEN

Dorsal raphe nucleus (DRN) neurons are reciprocally connected to the locus coeruleus (LC) and send neural pathways to the medial hypothalamus (MH). The aim of this work was to investigate whether the blockade of α1-, α2- or ß-noradrenergic receptors in the DRN or the inactivation of noradrenergic neurons in the LC modify defensive behaviours organised by MH neurons. For this purpose, Wistar male rats received microinjections of WB4101, RX821002, propranolol (α1-, α2- and ß-noradrenergic receptor antagonists, respectively) or physiological saline in the DRN, followed 10 min later by MH GABAA receptor blockade. Other groups of animals received DSP-4 (a noradrenergic neurotoxin), physiological saline or only a needle insertion (sham group) into the LC, and 5 days later, bicuculline or physiological saline was administered in the MH. In all these cases, after MH treatment, the frequency and duration of defensive responses were recorded over 15 min. An anterograde neural tract tracer was also deposited in the DRN. DRN neurons send pathways to lateral and dorsomedial hypothalamus. Blockade of α1- and ß-noradrenergic receptors in the DRN decreased escape reactions elicited by bicuculline microinjections in the MH. In addition, a significant increase in anxiety-like behaviours was observed after the blockade of α2-noradrenergic receptors in the DRN. LC pretreatment with DSP-4 decreased both anxiety- and panic attack-like behaviours evoked by GABAA receptor blockade in the MH. In summary, the present findings suggest that the norepinephrine-mediated system modulates defensive reactions organised by MH neurons at least in part via noradrenergic receptors recruitment on DRN neurons.


Asunto(s)
Núcleo Dorsal del Rafe/fisiología , Hipotálamo Medio/fisiología , Neuronas/fisiología , Pánico/fisiología , Receptores Adrenérgicos alfa/fisiología , Receptores Adrenérgicos beta/fisiología , Antagonistas Adrenérgicos alfa/administración & dosificación , Animales , Ansiedad/fisiopatología , Núcleo Dorsal del Rafe/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Pánico/efectos de los fármacos , Ratas Wistar
4.
Am J Physiol Endocrinol Metab ; 316(5): E948-E955, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30888861

RESUMEN

Increased GABAergic output in the ventromedial hypothalamus (VMH) contributes to counterregulatory failure in recurrently hypoglycemic (RH) rats, and lactate, an alternate fuel source in the brain, contributes to this phenomenon. The current study assessed whether recurring bouts of glucose deprivation enhanced neuronal lactate uptake and, if so, whether this influenced γ-aminobutyric acid (GABA) output and the counterregulatory responses. Glucose deprivation was induced using 5-thioglucose (5TG). Control rats received an infusion of artificial extracellular fluid. These groups were compared with RH animals. Subsequently, the rats underwent a hypoglycemic clamp with microdialysis. To test whether 5TG affected neuronal lactate utilization, a subgroup of 5TG-treated rats was microinjected with a lactate transporter inhibitor [cyano-4-hydroxycinnamate (4CIN)] just before the start of the clamp. Both RH and 5TG raised VMH GABA levels, and this was associated with impaired counterregulatory responses. 4CIN reduced VMH GABA levels and restored the hormone responses in the 5TG group. We then evaluated [14C]lactate uptake in hypothalamic neuronal cultures. Recurring exposure to low glucose increased monocarboxylate transporter-2 mRNA expression and augmented lactate uptake. Taken together, our data suggest that glucose deprivation, per se, enhances lactate utilization in hypothalamic neurons, and this may contribute to suppression of the counterregulatory responses to hypoglycemia.


Asunto(s)
Glucosa/metabolismo , Hipoglucemia/metabolismo , Hipotálamo Medio/citología , Ácido Láctico/metabolismo , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Radioisótopos de Carbono , Catecolaminas/metabolismo , Ácidos Cumáricos/farmacología , Glucosa/análogos & derivados , Glucosa/deficiencia , Glucosa/farmacología , Técnica de Clampeo de la Glucosa , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Microdiálisis , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/efectos de los fármacos , Transportadores de Ácidos Monocarboxílicos/genética , Neuronas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ácido gamma-Aminobutírico/efectos de los fármacos
5.
J Neuroendocrinol ; 30(11): e12642, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30168642

RESUMEN

The astrocytic glutamine (Gln)-glutamate (Glu) cycle (GGC) supplies Gln for the regulation of glutamatergic synaptic transmission (GST) in the adult hippocampus. Increased synaptic Glu release in the perinatal ventrolateral ventromedial nucleus of the hypothalamus (vlVMH) modulates sexual differentiation, however, whether GGC regulates GST in the perinatal vlVMH has not been determined. Sex differences in oestradiol (E2 ) levels exist in the neonatal hypothalamus, and E2 increases levels of glutamine synthetase and glutaminase, two key enzymes involved in the GGC. Thus, it is hypothesised that sexually dimorphic phenotypes may exist in glutamatergic synapses associated with the GGC in the vlVMH in perinatal rats. Whole-cell voltage-clamp recordings in vlVMH neurones in brain slices from male and female pups revealed that pharmacological disruption of the GGC by α-(methylamino) isobutyric acid (5 mmol L-1 ), which blocks neuronal Gln uptake; or by l-methionine sulphoximine (1.5 mmol L-1 ), which inhibits astrocytic Gln synthesis, decreased miniature excitatory postsynaptic current (mEPSC) amplitudes in female but not male pups. By contrast, GGC interruptions decreased evoked (e)EPSC amplitudes in both sexes following increased synaptic activity produced by a period of stimulation. In male pups, the decreased eEPSCs were attributable to reduced Glu release, as assessed by paired-pulse stimulations, whereas, in female pups, they were attributable to decreased Glu content in the synaptic vesicles, as measured by strontium-evoked mEPSCs. The l-methionine sulphoximine-mediated decrease in eEPSCs was rapidly rescued by exogenous Gln in female but not male pups. The reductions in mEPSCs and eEPSCs in female pups were accompanied by enhanced blocking effects of the low-affinity Glu AMPA receptor antagonist, γ-d-glutamylglycine, consistent with diminished Glu release. In conclusion, female, but not male pups, rely on constitutive astrocytic Gln for sustained synaptic Glu release in the vlVMH. This glutamatergic synaptic phenotype may be associated with brain and behaviour feminisation and/or defeminisation in rats.


Asunto(s)
Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Neuronas/fisiología , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Astrocitos/efectos de los fármacos , Potenciales Postsinápticos Excitadores , Femenino , Glutamina/administración & dosificación , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Masculino , Potenciales Postsinápticos Miniatura , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Caracteres Sexuales , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , beta-Alanina/administración & dosificación , beta-Alanina/análogos & derivados
6.
Brain Res ; 1650: 21-30, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566061

RESUMEN

The time course effects of ovarian steroids on kisspeptin and GnRH/LH systems is not totally clarified. We investigated the temporal relationship among kisspeptin and GnRH mRNA and kisspeptin content in the preoptic area (POA), GnRH content and release in the medial basal hypothalamus (MBH) and plasma LH levels under different steroid treatments. Ovariectomized rats treated with oil (OVOO), oil plus single dose of estradiol (OVOE), oil plus single dose of progesterone (OVOP), estradiol for 3 days plus oil (OVEO) or estradiol for 3 days plus progesterone (OVEP) were hourly decapitated from 10:00 to 17:00 or had the MBH microdialyzed from 09:00 to 19:00. Estradiol and progesterone acutely increased POA kisspeptin content without altering POA kisspeptin mRNA levels. Short-term exposure to both hormones stimulated MBH GnRH content, although no GnRH/LH surges had occurred. Chronic estradiol-treatment increased both kisspeptin mRNA levels and content in the POA, demonstrating that long exposure to estradiol is required to activate the whole kisspeptin synthesis machinery. This was followed by the peak in the GnRH/LH release. In estradiol-primed rats, progesterone further increased POA kisspeptin content, amplified and advanced GnRH/LH surges, with no additional change on POA kisspeptin mRNA. The data show an estradiol-induced temporal association between kisspeptin increase in the POA and GnRH/LH surges. Interestingly, the classic action of progesterone in amplifying and accelerating the GnRH/LH surges seems to occur by a mechanism which involves POA kisspeptin system.


Asunto(s)
Estradiol/metabolismo , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Animales , Estradiol/farmacología , Femenino , Gonadotropinas , Hipotálamo/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Kisspeptinas/efectos de los fármacos , Kisspeptinas/metabolismo , Hormona Luteinizante/sangre , Ovariectomía , Área Preóptica/efectos de los fármacos , Progesterona/metabolismo , Progesterona/farmacología , Ratas , Ratas Wistar , Análisis Espacio-Temporal
7.
Am J Physiol Regul Integr Comp Physiol ; 310(6): R476-80, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26739651

RESUMEN

Adropin, a recently described peptide hormone produced in the brain and liver, has been reported to have physiologically relevant actions on glucose homeostasis and lipogenesis, and to exert significant effect on endothelial function. We describe a central nervous system action of adropin to inhibit water drinking and identify a potential adropin receptor, the orphan G protein-coupled receptor, GPR19. Reduction in GPR19 mRNA levels in medial basal hypothalamus of male rats resulted in the loss of the inhibitory effect of adropin on water deprivation-induced thirst. The identification of a novel brain action of adropin and a candidate receptor for the peptide should extend and accelerate the study of the potential therapeutic value of adropin or its mimetics for the treatment of metabolic disorders.


Asunto(s)
Proteínas Sanguíneas/farmacología , Encéfalo/efectos de los fármacos , Conducta de Ingestión de Líquido/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Péptidos/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Neurotransmisores/efectos de los fármacos , Animales , Presión Arterial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Inyecciones Intraventriculares , Masculino , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotransmisores/metabolismo , Sed/efectos de los fármacos , Privación de Agua
8.
Neuroendocrinology ; 103(6): 711-23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26580201

RESUMEN

Substance P (SP) was recently reported to be expressed in human kisspeptin/neurokinin B/dynorphin (KNDy) neurons and to enhance KNDy neuron excitability in the mouse hypothalamus. We therefore examined (1) interactions of SP and kisspeptin in the mediobasal hypothalamus of adult male rhesus monkeys using immunofluorescence, and (2) the ability of SP to induce LH release in GnRH-primed, agonadal juvenile male monkeys. SP cell bodies were observed only occasionally in the arcuate nucleus (Arc), but more frequently dorsal to the Arc in the region of the premammillary nucleus. Castration resulted in an increase in the number of SP cell bodies in the Arc but not in the other regions. SP fibers innervated the Arc, where they were found in close apposition with kisspeptin perikarya in the periphery of this nucleus. Beaded SP axons projected to the median eminence, where they terminated in the external layer and intermingled with beaded kisspeptin axons. Colocalization of the two peptides, however, was not observed. Although close apposition between SP fibers and kisspeptin neurons suggest a role for SP in modulating GnRH pulse generator activity, i.v. injections of SP failed to elicit release of GnRH (as reflected by LH) in the juvenile monkey. Although the finding of structural interactions between SP and kisspeptin neurons is consistent with the notion that this tachykinin may be involved in regulating pulsatile GnRH release, the apparent absence of expression of SP in KNDy neurons suggests that this peptide is unlikely to be a fundamental component of the primate GnRH pulse generator.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Péptidos/administración & dosificación , Sustancia P/metabolismo , Administración Intravenosa , Animales , Castración , Relación Dosis-Respuesta a Droga , Hipotálamo Medio/citología , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Macaca mulatta , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo
9.
Brain Res Bull ; 118: 58-64, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26385088

RESUMEN

The RFamide peptide family comprises a number of biologically active peptides sharing RF motif at their C-terminal end. These peptides are involved in the control of multiple physiological functions including regulation of metabolism and feeding behavior. QRFP-43 as well as its 26-aminoacid residue QRFP-26 are able to cause orexigenic effect when administered to the rodents' cerebral ventricles. QRFPs have been suggested as the endogenous ligands of the previously orphan GPR103 receptors. GPR103 receptors share amino acid identity with other receptors of neuropeptides involved in feeding (NPY, NPFF, galanin). QRFP-26 expressing neurons and binding sites are densely present in the rat medial hypothalamus (MHA), an area directly responsible for the regulation of feeding. QRFP-26 was delivered to the target area by direct intrahypothalamic microinjection, and the consumption of liquid food was measured over a 60 min period. Both doses (100 and 200 ng) significantly increased food intake. Non-specific receptor antagonist BIBP3226 eliminated the orexigenic effect caused by QRFP-26 administration. Effective doses of QRFP-26 did not modify general locomotor activity and behavioral patterns examined in the open-field test. This study is the first reporting feeding modulating effects following direct intrahypothalamic QRFP-26 administration.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Péptidos/farmacología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Conducta Animal/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hipotálamo Medio/fisiología , Masculino , Microinyecciones , Actividad Motora/efectos de los fármacos , Ratas , Ratas Wistar
10.
Food Chem Toxicol ; 83: 10-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26032630

RESUMEN

Perfluorooctane sulfonate (PFOS) is a fluorinated organic compound. This chemical is neurotoxic and can alter the pituitary secretion. This is an initial study aimed at knowing the toxic effects of high doses of PFOS on prolactin secretion and the possible mechanisms involved in these alterations. For that, adult male rats were orally treated with 3.0 and 6.0 mg of PFOS/kg body weight (b.w.)/day for 28 days. At the end of the treatment, the serum levels of prolactin and estradiol as well as the concentration of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA) and gamma-aminobutyric acid (GABA) were quantified in the anterior and in the mediobasal hypothalamus. PFOS, at the administered doses, reduced prolactin and estradiol secretion, increased the concentration of dopamine and GABA in the anterior hypothalamus, and decreased the ratios DOPAC/dopamine and HVA/dopamine in this same hypothalamic area. The outcomes reported in this study suggest that (1) high doses of PFOS inhibit prolactin secretion in adult male rats; (2) only the periventricular-hypophysial dopaminergic (PHDA) neurons seem to be involved in this inhibitory effect but not the tuberoinfundibular dopaminergic (TIDA) and the tuberohypophysial dopaminergic (THDA) systems; (3) GABAergic cells from the paraventricular and supraoptic nuclei could be partially responsible for the PFOS action on prolactin secretion; and finally (4) estradiol might take part in the inhibition exerted by elevated concentration of PFOS on prolactin release.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Hipotálamo Anterior/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Prolactina/antagonistas & inhibidores , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Dopamina/química , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/administración & dosificación , Contaminantes Ambientales/administración & dosificación , Estradiol/sangre , Estradiol/metabolismo , Fluorocarburos/administración & dosificación , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Hipotálamo Anterior/metabolismo , Hipotálamo Medio/metabolismo , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Adenohipófisis/metabolismo , Prolactina/sangre , Prolactina/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo , Pruebas de Toxicidad Subaguda , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/metabolismo
11.
J Alzheimers Dis ; 46(4): 843-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25869787

RESUMEN

Patients with Alzheimer's disease (AD) have a higher risk for developing insulin resistance and diabetes. Amyloid plaques, a hallmark of AD, are composed of amyloid-ß (Aß). Because the mediobasal hypothalamus controls hepatic glucose production, we examined the hypothesis that its exposure to Aß perturbs the regulation of glucose metabolism. The infusion of Aß25-35, but not its scrambled counterpart, into the mediobasal hypothalamus of young rats, increased circulating glucose as a consequence of enhanced hepatic glucose production during pancreatic clamp studies. These findings suggest a link between AD and alterations of glucose metabolism.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Glucemia/metabolismo , Hipotálamo Medio/efectos de los fármacos , Hígado/metabolismo , Fragmentos de Péptidos/farmacología , Animales , Técnica de Clampeo de la Glucosa , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/efectos de los fármacos , Masculino , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Brain Res ; 1608: 75-81, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25725382

RESUMEN

The contribution of exercise to hyperthermia mediated by MDMA is not known. We recently showed that inhibiting the dorsomedial hypothalamus (DMH) attenuated spontaneous locomotion and hyperthermia and prevented deaths in rats given MDMA in a warm environment. The goal of this study was to confirm that restoring locomotion through a treadmill would reverse these effects thereby confirming that locomotion mediated by the DMH contributes to MDMA-mediated hyperthermia. Rats were randomized to receive bilateral microinjections, into the region of the DMH, of muscimol (80pmol/100nl) or artificial CSF followed by a systemic dose of either MDMA (7.5mg/kg, i.v.) or saline. Immediately after the systemic injection, rats were placed on a motorized treadmill maintained at 32°C. Rats were exercised at a fixed speed (10m/min) until their core temperature reached 41°C. Our results showed that a fixed exercise load abolished the decreases in temperature and mortality, seen previously with inhibition of the DMH in freely moving rats. Therefore, locomotion mediated by neurons in the DMH is critical to the development of hyperthermia from MDMA.


Asunto(s)
Prueba de Esfuerzo/efectos adversos , Fiebre/etiología , Alucinógenos/farmacología , Hipotálamo Medio/efectos de los fármacos , Locomoción/efectos de los fármacos , N-Metil-3,4-metilenodioxianfetamina/farmacología , Análisis de Varianza , Animales , Temperatura Corporal/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Masculino , Microinyecciones , Muscimol/farmacología , Ratas , Ratas Sprague-Dawley , Telemetría
13.
Alcohol Clin Exp Res ; 38(10): 2572-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25335926

RESUMEN

BACKGROUND: Insulin-like growth factor-1 (IGF-1) and transforming growth factor ß1 (TGFß1) are produced in hypothalamic astrocytes and facilitate luteinizing hormone-releasing hormone (LHRH) secretion. IGF-1 stimulates release by acting directly on the LHRH nerve terminals and both peptides act indirectly through specific plastic changes on glial/tanycyte processes that further support LHRH secretion. Because the relationship between these growth factors in the hypothalamus is not known, we assessed the ability of IGF-1 to induce TGFß1 synthesis and release and the actions of alcohol (ALC) on this mechanism prior to the onset of puberty. METHODS: Hypothalamic astrocytes were exposed to medium only, medium plus IGF-1 (200 ng/ml), or medium plus IGF-1 with 50 mM ALC. After 18 hours, media were collected and assayed for TGFß1. For the in vivo experiment, prepubertal female rats were administered either ALC (3 g/kg) or water via gastric gavage at 07:30 hours and at 11:30 hours. At 09:00 hours, saline or IGF-1 was administered into the third ventricle. Rats were killed at 15:00 hours and the medial basal hypothalamus (MBH) was collected for assessment of TGFß1, IGF-1 receptor (IGF-1R), and Akt. RESULTS: IGF-1 induced TGFß1 release (p < 0.01) from hypothalamic astrocytes in culture, an action blocked by ALC. In vivo, IGF-1 administration caused an increase in TGFß1 protein compared with controls (p < 0.05), an action blocked by ALC as well as a phosphatidylinositol 3 kinase/Akt inhibitor. IGF-1 stimulation also increased both total (p< 0.01) and phosphorylated (p)-IGF-1R (p < 0.05) protein levels, and phosphorylated (p)-Akt levels (p < 0.01), which were also blocked by ALC. CONCLUSIONS: This study shows that ALC blocks IGF-1 actions to stimulate synthesis and release of hypothalamic TGFß1, total and p-IGF-1R, and p-Akt levels further demonstrating the inhibitory actions of ALC on puberty-related events associated with hypothalamic LHRH release.


Asunto(s)
Etanol/farmacología , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Maduración Sexual , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio/citología , Técnicas In Vitro , Modelos Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/metabolismo
14.
Alcohol Clin Exp Res ; 38(5): 1321-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24588206

RESUMEN

BACKGROUND: Alcohol (ALC) diminishes gonadotropin-releasing hormone (GnRH) secretion and delays puberty. Glial transforming growth factor ß1 (TGFß1) plays a role in glial-neuronal communications facilitating prepubertal GnRH secretion. We assessed the effects of acute ALC administration on TGFß1-induced GnRH gene expression in the brain preoptic area (POA) and release of the peptide from the medial basal hypothalamus (MBH). Furthermore, we assessed actions and interactions of TGFß1 and ALC on an adhesion/signaling gene family involved in glial-neuronal communications. METHODS: Prepubertal female rats were administered ALC or water via gastric gavage at 7:30 am. At 9:00 am, saline or TGFß1 (100 ng/3 µl) was administered into the third ventricle. At 3:00 pm, the POA was removed and frozen for gene expression analysis and repeated for protein assessments. In another experiment, the MBH was removed from ALC-free rats. After equilibration, tissues were incubated in Locke's medium only or medium containing TGFß1 with or without 50 mM ALC for measurement of GnRH peptide released in vitro. RESULTS: TGFß1 induced GnRH gene expression in the POA, and this effect was blocked by ALC. We also described the presence and responsiveness of the TGFß1 receptor in the POA and showed that acute ALC exposure not only altered the TGFß1-induced increase in TGFß-R1 protein expression but also the activation of receptor-associated proteins, Smad2 and Smad3, key downstream components of the TGFß1 signaling pathway. Assessment of an adhesion/signaling family consisting of glial receptor protein tyrosine phosphatase beta and neuronal contactin-associated protein-1 (Caspr1) and contactin showed that the neuronal components were induced by TGFß1 and that ALC blocked these effects. Finally, TGFß1 was shown to induce release of the GnRH peptide in vitro, an action that was blocked by ALC. CONCLUSIONS: We have demonstrated glial-derived TGFß1 induces GnRH gene expression in the POA and stimulates release of the peptide from the MBH, actions necessary for driving the pubertal process. Importantly, ALC acted at both brain regions to block stimulatory effects of TGFß1. Furthermore, ALC altered neuronal components of an adhesion/signaling family previously shown to be expressed on GnRH neurons and implicated in glial-GnRH neuronal communications. These results further demonstrate detrimental effects of ALC at puberty.


Asunto(s)
Etanol/farmacología , Hormona Liberadora de Gonadotropina/biosíntesis , Hipotálamo Medio/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Interacciones Farmacológicas , Femenino , Expresión Génica/efectos de los fármacos , Hipotálamo Medio/metabolismo , Área Preóptica/metabolismo , Pubertad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Endocrinology ; 155(5): 1874-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24484170

RESUMEN

Kisspeptin regulates reproductive events, including puberty and ovulation, primarily via GnRH neurons. Prolonged treatment of prepubertal striped bass females with kisspeptin (Kiss) 1 or Kiss2 peptides failed to enhance puberty but suggested a gnrh-independent pituitary control pathway. Kiss2 inhibited, but Kiss1 stimulated, FShß expression and gonadal development, although hypophysiotropic gnrh1 and gnrh receptor expression remained unchanged. In situ hybridization and immunohistochemistry on brains and pituitaries revealed a differential plasticity between the 2 kisspeptin neurons. The differences were most pronounced at the prespawning phase in 2 regions along the path of gnrh1 axons: the nucleus lateralis tuberis (NLT) and the neurohypophysis. Kiss1 neurons appeared in the NLT and innervated the neurohypophysis of prespawning males and females, reaching Lh gonadotropes in the proximal pars distalis. Males, at all reproductive stages, had Kiss2 innervations in the NLT and the neurohypophysis, forming large axonal bundles in the former and intermingling with gnrh1 axons. Unlike in males, only preovulatory females had massive NLT-neurohypophysis staining of kiss2. Kiss2 neurons showed a distinct appearance in the NLT pars ventralis-equivalent region only in spawning zebrafish, indicating that this phenomenon is widespread. These results underscore the NLT as important nuclei for kisspeptin action in 2 facets: 1) kisspeptin-gnrh interaction, both kisspeptins are involved in the regulation of gnrh release, in a stage- and sex-dependent manner, especially at the prespawning phase; and 2) gnrh-independent effect of Kiss peptides on the pituitary, which together with the plastic nature of their neuronal projections to the pituitary implies that a direct gonadotropic regulation is plausible.


Asunto(s)
Lubina/fisiología , Proteínas de Peces/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas/metabolismo , Maduración Sexual , Proteínas de Xenopus/metabolismo , Animales , Acuicultura , Axones/efectos de los fármacos , Axones/metabolismo , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Femenino , Fármacos para la Fertilidad Femenina/farmacología , Proteínas de Peces/biosíntesis , Proteínas de Peces/genética , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/genética , Sistema Hipotálamo-Hipofisario/citología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Hipotálamo Medio/citología , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/crecimiento & desarrollo , Hipotálamo Medio/metabolismo , Kisspeptinas/administración & dosificación , Kisspeptinas/farmacología , Maryland , Neurohipófisis/citología , Neurohipófisis/efectos de los fármacos , Neurohipófisis/crecimiento & desarrollo , Neurohipófisis/metabolismo , Maduración Sexual/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas de Xenopus/administración & dosificación , Proteínas de Xenopus/farmacología
16.
Cereb Cortex ; 24(6): 1518-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23349224

RESUMEN

The aim of the present study was to investigate the involvement of N-methyl-d-aspartate (NMDA) and amino-3-hydroxy-5-methyl-isoxazole-4-proprionate (AMPA)/kainate receptors of the prelimbic (PL) division of the medial prefrontal cortex (MPFC) on the panic attack-like reactions evoked by γ-aminobutyric acid-A receptor blockade in the medial hypothalamus (MH). Rats were pretreated with NaCl 0.9%, LY235959 (NMDA receptor antagonist), and NBQX (AMPA/kainate receptor antagonist) in the PL at 3 different concentrations. Ten minutes later, the MH was treated with bicuculline, and the defensive responses were recorded for 10 min. The antagonism of NMDA receptors in the PL decreased the frequency and duration of all defensive behaviors evoked by the stimulation of the MH and reduced the innate fear-induced antinociception. However, the pretreatment of the PL cortex with NBQX was able to decrease only part of defensive responses and innate fear-induced antinociception. The present findings suggest that the NMDA-glutamatergic system of the PL is critically involved in panic-like responses and innate fear-induced antinociception and those AMPA/kainate receptors are also recruited during the elaboration of fear-induced antinociception and in panic attack-related response. The activation of the glutamatergic neurotransmission of PL division of the MPFC during the elaboration of oriented behavioral reactions elicited by the chemical stimulation of the MH recruits mainly NMDA receptors in comparison with AMPA/kainate receptors.


Asunto(s)
Conducta Animal/fisiología , Miedo/fisiología , Hipotálamo Medio/fisiología , Percepción del Dolor/fisiología , Pánico/fisiología , Corteza Prefrontal/fisiología , Animales , Conducta Animal/efectos de los fármacos , Bicuculina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Miedo/efectos de los fármacos , Antagonistas de Receptores de GABA-A/farmacología , Hipotálamo Medio/efectos de los fármacos , Isoquinolinas/farmacología , Masculino , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/fisiopatología , Percepción del Dolor/efectos de los fármacos , Pánico/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Quinoxalinas/farmacología , Ratas Wistar , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
17.
J Neurosci ; 33(49): 19051-9, 2013 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-24305803

RESUMEN

Release of gonadotropin releasing hormone (GnRH) from the medial basal hypothalamus (MBH)/median eminence region (S-ME) is essential for normal reproductive function. GnRH release is profoundly regulated by the negative and positive feedback effects of ovarian estradiol (E2). Here we report that neuroestradiol, released in the S-ME, also directly influences GnRH release in ovariectomized female monkeys, in which the ovarian source of E2 is removed. We found that (1) brief infusion of E2 benzoate (EB) to the S-ME rapidly stimulated release of GnRH and E2 in the S-ME of ovariectomized monkeys, (2) electrical stimulation of the MBH resulted in GnRH release as well as E2 release, and (3) direct infusion of an aromatase inhibitor to the S-ME suppressed spontaneous GnRH release as well as the EB-induced release of GnRH and E2. These findings reveal the importance of neuroestradiol as a neurotransmitter in regulation of GnRH release. How circulating ovarian E2 interacts with hypothalamic neuroestrogens in the control of GnRH release remains to be investigated.


Asunto(s)
Estradiol/análogos & derivados , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Animales , Inhibidores de la Aromatasa/farmacología , Cromatografía Líquida de Alta Presión , Estimulación Eléctrica , Electrodos Implantados , Estradiol/farmacología , Femenino , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Letrozol , Macaca mulatta , Espectrometría de Masas , Eminencia Media/efectos de los fármacos , Eminencia Media/metabolismo , Microdiálisis , Nitrilos/farmacología , Ovariectomía , Radioinmunoensayo , Triazoles/farmacología
18.
Endocrinology ; 154(9): 3130-40, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23751875

RESUMEN

Chronic inflammation is involved in the pathogenesis of obesity and type 2 diabetes. Recently teasaponin, an extract from tea, has been shown to have antiinflammatory effects. We examined the effect of teasaponin on obesity, inflammation, glucose metabolism, and central leptin sensitivity in obese mice fed a high-fat (HF) diet for 16 weeks. Intraperitoneal injections of teasaponin (10 mg/kg, daily) for 21 days significantly decreased the food intake and body weight of HF diet-induced obese mice. Teasaponin treatment also reduced the protein levels of proinflammatory cytokines (TNF-α, IL-6, and/or IL-1ß) and nuclear factor-κB signaling (phosphorylated inhibitory-κB kinase and phosphorylated inhibitory-κBα) in adipose tissue and the liver. The antiinflammatory effects of teasaponin were associated with improved glycemic status in the treated animals, evidenced by improved glucose tolerance, homeostasis model assessment, and fasting plasma insulin. In the hypothalamus, teasaponin decreased both proinflammatory cytokines and inflammatory signaling in the mediobasal hypothalamus. Teasaponin treatment also enhanced the anorexigenic effect of central leptin administration, restored leptin phosphorylated signal transducer and activator of transcription-3 (p-STAT3) signaling in the arcuate nucleus, and increased hypothalamic expression of the anorexigenic peptide proopiomelanocortin. These results identify a potential novel application for teasaponin as an antiobesity and antiinflammatory agent.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Camellia sinensis/química , Hipotálamo Medio/efectos de los fármacos , Leptina/metabolismo , Obesidad/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Té/química , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Depresores del Apetito/administración & dosificación , Depresores del Apetito/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Hipotálamo Medio/inmunología , Hipotálamo Medio/metabolismo , Inyecciones Intraperitoneales , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , Extractos Vegetales/administración & dosificación , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos
19.
Neuropharmacology ; 71: 27-36, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23542442

RESUMEN

Hemopressin is the first peptide ligand to be described for the CB1 cannabinoid receptor. Hemopressin acts as an inverse agonist in vivo and can cross the blood-brain barrier to both inhibit appetite and induce antinociception. Despite being highly effective, synthetic CB1 inverse agonists are limited therapeutically due to unwanted, over dampening of central reward pathways. However, hemopressin appears to have its effect on appetite by affecting satiety rather than reward, suggesting an alternative mode of action which might avoid adverse side effects. Here, to resolve the neuronal circuitry mediating hemopressin's actions, we have combined blood-oxygen-level-dependent, pharmacological-challenge magnetic resonance imaging with c-Fos functional activity mapping to compare brain regions responsive to systemic administration of hemopressin and the synthetic CB1 inverse agonist, AM251. Using these complementary methods, we demonstrate that hemopressin activates distinct neuronal substrates within the brain, focused mainly on the feeding-related circuits of the mediobasal hypothalamus and in nociceptive regions of the periaqueductal grey (PAG) and dorsal raphe (DR). In contrast to AM251, there is a distinct lack of activation of the brain reward centres, such as the ventral tegmental area, nucleus accumbens and orbitofrontal cortex, which normally form a functional activity signature for the central action of synthetic CB1 receptor inverse agonists. Thus, hemopressin modulates the function of key feeding-related brain nuclei of the mediobasal hypothalamus, and descending pain pathways of the PAG and DR, and not higher limbic structures. Thus, hemopressin may offer behaviourally selective effects on nociception and appetite, without engaging reward pathways.


Asunto(s)
Depresores del Apetito/farmacología , Hemoglobinas/farmacología , Hipotálamo Medio/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Sustancia Gris Periacueductal/efectos de los fármacos , Núcleos del Rafe/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/efectos adversos , Conducta Animal/efectos de los fármacos , Cannabinoides/administración & dosificación , Cannabinoides/efectos adversos , Cannabinoides/farmacología , Hemoglobinas/administración & dosificación , Hemoglobinas/efectos adversos , Hipotálamo Medio/metabolismo , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/efectos adversos , Sustancia Gris Periacueductal/citología , Sustancia Gris Periacueductal/metabolismo , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Piperidinas/farmacología , Pirazoles/administración & dosificación , Pirazoles/efectos adversos , Pirazoles/farmacología , Distribución Aleatoria , Núcleos del Rafe/citología , Núcleos del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Respuesta de Saciedad/efectos de los fármacos
20.
Physiol Behav ; 110-111: 158-68, 2013 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-23313561

RESUMEN

Increasing estradiol concentrations during the late follicular phase stimulate sexual behavior and the GnRH/LH surge, and it is known that kisspeptin signaling is essential for the latter. Administration of LPS can block these events, but the mechanism involved is unclear. We examined brain tissue from intact ewes to determine: i) which regions are activated with respect to sexual behavior, the LH surge and LPS administration, ii) the location and activation pattern of kisspeptin cells in control and LPS treated animals, and iii) whether CRFR type 2 is involved in such disruptive mechanisms. Follicular phases were synchronized with progesterone vaginal pessaries and control animals were killed at 0 h, 16 h, 31 h or 40 h (n=4-6/group) after progesterone withdrawal (time zero). At 28 h, other animals received endotoxin (LPS; 100 ng/kg) and were subsequently killed at 31 h or 40 h (n=5/group). LH surges only occurred in control ewes, during which there was a marked increase in c-Fos expression within the ventromedial nucleus (VMN), arcuate nucleus (ARC), and medial preoptic area (mPOA), as well as an increase in the percentage of kisspeptin cells co-expressing c-Fos in the ARC and mPOA compared to animals sacrificed at all other times. Expression of c-Fos also increased in the bed nucleus of the stria terminalis (BNST) in animals just before the expected onset of sexual behavior. However, LPS treatment increased c-Fos expression within the VMN, ARC, mPOA and diagonal band of broca (dBb), along with CRFR type 2 immunoreactivity in the lower part of the ARC and median eminence (ME), compared to controls. Furthermore, the percentage of kisspeptin cells co-expressing c-Fos was lower in the ARC and mPOA. Thus, we hypothesize that in intact ewes, the BNST is involved in the initiation of sexual behavior while the VMN, ARC, and mPOA as well as kisspeptin cells located in the latter two areas are involved in estradiol positive feedback only during the LH surge. By contrast, disruption of sexual behavior and the LH surge after LPS involves cells located in the VMN, ARC, mPOA and dBb, as well as cells containing CRFR type 2 in the lower part of the ARC and ME, and is accompanied by inhibition of kisspeptin cell activation in both the ARC and mPOA.


Asunto(s)
Ciclo Estral/fisiología , Genes fos/genética , Hipotálamo Medio/efectos de los fármacos , Hipotálamo Medio/metabolismo , Kisspeptinas/genética , Lipopolisacáridos/farmacología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Interpretación Estadística de Datos , Estradiol/sangre , Ciclo Estral/efectos de los fármacos , Femenino , Inmunohistoquímica , Kisspeptinas/biosíntesis , Hormona Luteinizante/sangre , Masculino , Progesterona/sangre , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Núcleos Septales/metabolismo , Conducta Sexual Animal/fisiología , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA