Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
Domest Anim Endocrinol ; 74: 106486, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32882449

RESUMEN

Gonadotropin inhibitory hormone (GnIH), initially discovered in birds as a hypothalamic neuropeptide, inhibits the synthesis and release of gonadotropins by affecting GnRH neurons and gonadotropes. Therefore, it may be a key neuropeptide in reproduction in birds. The aim of the present study was to investigate the prepubertal, pubertal, and postpubertal localization of GnIH and changes in hypothalamic GnIH expression in British United Turkey hens. In prepubertal, pubertal, and postpubertal periods, the brains of turkey hens (n = 15) were removed after fixation. Sections (30 µm) were prepared from the entire hypothalamus and stained immunohistochemically against GnIH antibody. Gonadotropin inhibitory hormone-immunoreactive neurons were observed in the paraventricular nucleus. These neurons were significantly more abundant in the prepubertal turkeys than pubertal and postpubertal turkeys (P < 0.05). The results suggested that GnIH neurons have an important role in regulating the pubertal events in British United Turkey hens.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Maduración Sexual/fisiología , Pavos/fisiología , Animales , Femenino , Hormonas Hipotalámicas/genética , Neuronas/clasificación , Neuronas/fisiología
2.
Orphanet J Rare Dis ; 15(1): 269, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32993765

RESUMEN

STUDY OBJECTIVES: Niemann-Pick type C (NPC) is an autosomal recessive and congenital neurological disorder characterized by the accumulation of cholesterol and glycosphingolipids. Symptoms include hepatosplenomegaly, vertical supranuclear saccadic palsy, ataxia, dystonia, and dementia. Some cases frequently display narcolepsy-like symptoms, including cataplexy which was reported in 26% of all NPC patients and was more often recorded among late-infantile onset (50%) and juvenile onset (38%) patients. In this current study, we examined CSF orexin levels in the 10 patients of NPC with and without cataplexy, which supports previous findings. METHODS: Ten patients with NPC were included in the study (5 males and 5 females). NPC diagnosis was biochemically confirmed in all 10 patients, from which 8 patients with NPC1 gene were identified. We compared CSF orexin levels among NPC, narcoleptic and idiopathic hypersomnia patients. RESULTS: Six NPC patients with cataplexy had low or intermediate orexin levels. In 4 cases without cataplexy, their orexin levels were normal. In 5 cases with Miglustat treatment, their symptoms stabilized or improved. For cases without Miglustat treatment, their conditions worsened generally. The CSF orexin levels of NPC patients were significantly higher than those of patients with narcolepsy-cataplexy and lower than those of patients with idiopathic hypersomnia, which was considered as the control group with normal CSF orexin levels. DISCUSSION: Our study indicates that orexin level measurements can be an early alert of potential NPC. Low or intermediate orexin levels could further decrease due to reduction in the neuronal function in the orexin system, accelerating the patients' NPC pathophysiology. However with Miglustat treatment, the orexin levels stabilized or improved, along with other general symptoms. Although the circuitry is unclear, this supports that orexin system is indeed involved in narcolepsy-cataplexy in NPC patients. CONCLUSION: The NPC patients with cataplexy had low or intermediate orexin levels. In the cases without cataplexy, their orexin levels were normal. Our study suggests that orexin measurements can serve as an early alert for potential NPC; furthermore, they could be a marker of therapy monitoring during a treatment.


Asunto(s)
Cataplejía , Enfermedad de Niemann-Pick Tipo C , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/uso terapéutico , Cataplejía/tratamiento farmacológico , Femenino , Humanos , Masculino , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Orexinas
3.
J Neuroendocrinol ; 30(7): e12597, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29624758

RESUMEN

At the turn of the millennium, a neuropeptide with pronounced inhibitory actions on avian pituitary gonadotrophin secretion was identified and named gonadotrophin-inhibitory hormone (GnIH). Across bird species, GnIH acts at the level of the pituitary and the gonadotrophin-releasing hormone (GnRH) neuronal system to inhibit reproduction. Subsequent to this initial discovery, orthologues of GnIH have been identified and characterised across a broad range of species. In many vertebrates, the actions of GnIH and its orthologues serve functional roles analogous to those seen in birds. In other cases, GnIH and its orthologues exhibit more diverse actions dependent on sex, species, season and reproductive condition. The present review highlights the discovery and functional implications of GnIH across species, focusing on research domains in which the significance of this neuropeptide has been explored most.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Reproducción/fisiología , Estrés Psicológico/metabolismo , Animales , Aves , Mamíferos
4.
J Neuroendocrinol ; 29(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28683170

RESUMEN

Gonadotrophin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotrophin synthesis and release in birds and mammals. In Japanese quail, GnIH neurones express the noradrenergic receptor and receive noradrenergic innervation. Treatment with noradrenaline (NA) stimulates GnIH release from diencephalic tissue blocks in vitro. However, the effects of NA on hypothalamic GnIH gene expression have not been determined. We investigated noradrenergic regulation of GnIH gene expression in the brain of male quail using the selective noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4). We first showed that DSP-4 reduced the number of noradrenergic (dopamine-ß-hydroxylase immunoreactive) cells in the locus coeruleus (LoC) and specifically lowered the NA concentration in the hypothalamus of male quail. Other monoamines, such as dopamine and serotonin, were not affected by drug treatment. DSP-4 did not decrease the numbers of noradrenergic cells of the lateral tegmental cell group, nor the plasma NA concentration. Decreased hypothalamic NA levels after DSP-4 treatment did not change GnIH gene expression in the brains of quail during their interaction with conspecifics. On the other hand, GnIH gene expression increased in the brains of quail socially isolated for 1 hour after DSP-4 treatment. These results suggest that some noradrenergic neurones have inhibitory effects on GnIH gene expression of the hypothalamus in solitary quail.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Proteínas Aviares/genética , Coturnix , Hormonas Hipotalámicas/genética , Hipotálamo/metabolismo , Animales , Bencilaminas/administración & dosificación , Glucemia/metabolismo , Diencéfalo/efectos de los fármacos , Diencéfalo/metabolismo , Expresión Génica , Hipotálamo/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Norepinefrina/sangre , ARN Mensajero/metabolismo , Aislamiento Social
5.
J Neuroendocrinol ; 28(5)2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26919074

RESUMEN

The role of gonadotrophin-inhibitory hormone (GnIH) in the inhibition of the reproductive axis has been well-established in birds and mammals. However, its role in other vertebrates, such as the teleost fish, remains controversial. In this context, the present study aimed to evaluate whether GnIH modulates the release of gonadotrophins and growth hormone (GH) in the cichlid fish Cichlasoma dimerus. First, we partially sequenced the precursor polypeptide for GnIH and identified three putative GnIH peptides. Next, we analysed the expression of this precursor polypeptide via a polymerase chain reaction in the reproductive axis of both sexes. We found a high expression of the polypeptide in the hypothalamus and gonads of males. Immunocytochemistry allowed the observation of GnIH-immunoreactive somata in the nucleus posterioris periventricularis and the nucleus olfacto-retinalis, with no differences between the sexes. GnIH-immunoreactive fibres were present in all brain regions, with a high density in the nucleus lateralis tuberis and at both sides of the third ventricle. Finally, we performed in vitro studies on intact pituitary cultures to evaluate the effect of two doses (10(-6)  m and 10(-8)  m) of synthetic C. dimerus (cd-) LPQRFa-1 and LPQRFa-2 on the release of gonadotrophins and GH. We observed that cd-LPQRFa-1 decreased ß-luteinising hormone (LH) and ß-follicle-stimulating hormone (FSH) and also increased GH release to the culture medium. The release of ß-FSH was increased only when it was stimulated with the higher cd-LPQRFa-2 dose. The results of the present study indicate that cd-LPQRFa-1, the cichlid fish GnIH, inhibits ß-LH and ß-FSH release and stimulates GH release in intact pituitary cultures of C. dimerus. The results also show that cd-LPQRF-2 could act as an ß-FSH-releasing factor in this fish species.


Asunto(s)
Cíclidos/metabolismo , Proteínas de Peces/metabolismo , Gonadotropinas/metabolismo , Hormona del Crecimiento/metabolismo , Hormonas Hipotalámicas/metabolismo , Animales , Cíclidos/genética , Femenino , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormonas Hipotalámicas/análisis , Hormonas Hipotalámicas/genética , Masculino , Hormonas Peptídicas/administración & dosificación , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo
6.
Gen Comp Endocrinol ; 227: 16-26, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26474923

RESUMEN

Seasonal breeding is widespread in vertebrates and involves sequential development of the gonads, onset of breeding activities (e.g. cycling in females) and then termination resulting in regression of the reproductive system. Whereas males generally show complete spermatogenesis prior to and after onset of breeding, females of many vertebrate species show only partial ovarian development and may delay onset of cycling (e.g. estrous), yolk deposition or germinal vesicle breakdown until conditions conducive for ovulation and onset of breeding are favorable. Regulation of this "brake" on the onset of breeding remains relatively unknown, but could have profound implications for conservation efforts and for "mismatches" of breeding in relation to global climate change. Using avian models it is proposed that a brain peptide, gonadotropin-inhibitory hormone (GnIH), may be the brake to prevent onset of breeding in females. Evidence to date suggests that although GnIH may be involved in the regulation of gonadal development and regression, it plays more regulatory roles in the process of final ovarian development leading to ovulation, transitions from sexual to parental behavior and suppression of reproductive function by environmental stress. Accumulating experimental evidence strongly suggests that GnIH inhibits actions of gonadotropin-releasing hormones on behavior (central effects), gonadotropin secretion (central and hypophysiotropic effects), and has direct actions in the gonad to inhibit steroidogenesis. Thus, actual onset of breeding activities leading to ovulation may involve environmental cues releasing an inhibition (brake) on the hypothalamo-pituitary-gonad axis.


Asunto(s)
Cambio Climático , Hormonas Hipotalámicas/metabolismo , Reproducción/fisiología , Pájaros Cantores/fisiología , Animales , Proteínas Aviares/metabolismo , Estro/fisiología , Femenino , Gónadas/metabolismo , Masculino , Estaciones del Año , Espermatogénesis/fisiología
7.
J Neuroendocrinol ; 27(6): 556-66, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25494813

RESUMEN

In 2000, we discovered a novel hypothalamic neuropeptide that actively inhibits gonadotrophin release in quail and termed it gonadotrophin-inhibitory hormone (GnIH). GnIH peptides have subsequently been identified in most representative species of gnathostomes. They all share a C-terminal LPXRFamide (X = L or Q) motif. GnIH can inhibit gonadotrophin synthesis and release by decreasing the activity of GnRH neuroes, as well as by directly inhibiting pituitary gonadotrophin secretion in birds and mammals. To investigate the evolutionary origin of GnIH and its ancestral function, we identified a GnIH precursor gene encoding GnIHs from the brain of sea lamprey, the most ancient lineage of vertebrates. Lamprey GnIHs possess a C-terminal PQRFamide motif. In vivo administration of one of lamprey GnIHs stimulated the expression of lamprey GnRH in the hypothalamus and gonadotophin ß mRNA in the pituitary. Thus, GnIH may have emerged in agnathans as a stimulatory neuropeptide that subsequently diverged to an inhibitory neuropeptide during the course of evolution from basal vertebrates to later-evolved vertebrates, such as birds and mammals. From a structural point of view, pain modulatory neuropeptides, such as neuropeptide FF (NPFF) and neuropeptide AF, share a C-terminal PQRFamide motif. Because agnathans possess both GnIH and NPFF genes, the origin of GnIH and NPFF genes may date back before the emergence of agnathans. More recently, we identified a novel gene encoding RFamide peptides in the amphioxus. Molecular phylogenetic analysis and synteny analysis indicated that this gene is closely related to the genes of GnIH and NPFF of vertebrates. The results suggest that the identified protochordate gene is similar to the common ancestor of GnIH and NPFF genes, indicating that the origin of GnIH and NPFF may date back to the time of the emergence of early chordates. The GnIH and NPFF genes may have diverged by whole-genome duplication during the course of vertebrate evolution.


Asunto(s)
Evolución Biológica , Gonadotropinas/fisiología , Hormonas Hipotalámicas/genética , Secuencia de Aminoácidos , Animales , Cordados , Humanos , Hormonas Hipotalámicas/química , Hormonas Hipotalámicas/fisiología , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
8.
J Neuroendocrinol ; 26(3): 186-94, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24528197

RESUMEN

Gonadotrophin-inhibitory hormone (GnIH) is a novel hypothalamic neuropeptide that was discovered in birds as an inhibitory factor for gonadotrophin release. RFamide-related peptide (RFRP) is a mammalian GnIH orthologue that inhibits gonadotrophin synthesis and release in mammals through actions on gonadotrophin-releasing hormone (GnRH) neurones and gonadotrophs, mediated via the GnIH receptor (GnIH-R), GPR147. On the other hand, hypothalamic kisspeptin provokes the release of GnRH from the hypothalamus. The present study aimed to compare the expression of RFRP in the dorsomedial hypothalamus and paraventricular nucleus (DMH/PVN) and that of kisspeptin in the arcuate nucleus (ARC) of the female goat hypothalamus during anoestrous and breeding seasons. Mature female Abadeh does were used during anoestrus, as well as the follicular and luteal phases of the cycle. The number of RFRP-immunoreactive (-IR) neurones in the follicular phase was lower than in the luteal and anoestrous stages. Irrespective of the ovarian stage, the number of RFRP-IR neurones in the rostral and middle regions of the DMH/PVN was higher than in the caudal region. By contrast, the number of kisspeptin-IR neurones in the follicular stage was greater than in the luteal stage and during the anoestrous stage. Irrespective of the stage of the ovarian cycle, the number of kisspeptin-IR neurones in the caudal region of the ARC was greater than in the middle and rostral regions. In conclusion, RFRP-IR cells were more abundant in the rostral region of the DMH/PVN nuclei of the hypothalamus, with a greater number being found during the luteal and anoestrous stages compared to the follicular stage. On the other hand, kisspeptin-IR neurones were more abundant in the caudal part of the ARC, with a greater number recorded in the follicular stage compared to the luteal and anoestrous stages.


Asunto(s)
Estro , Cabras/fisiología , Gonadotropinas/antagonistas & inhibidores , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuropéptidos/metabolismo , Reproducción , Animales , Femenino , Gonadotropinas/metabolismo
9.
J Neuroendocrinol ; 23(9): 791-803, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21696471

RESUMEN

Several neuropeptides with the C-terminal Arg-Phe-NH(2) (RFa) sequence have been identified in the hypothalamus of a variety of vertebrates. The present study was conducted to isolate novel RFa peptides from the zebra finch brain. Peptides were isolated by immunoaffinity purification using an antibody that recognises avian RFa peptides. The isolated peptide consisted of 25 amino acids with RFa at its C-terminus. The sequence was SGTLGNLAEEINGYNRRKGGFTFRFa. Alignment of the peptide with vertebrate 26RFa has revealed that the identified peptide is the zebra finch 26RFa. We also cloned the precursor cDNA encoding this peptide. Synteny analysis of the gene showed a high conservation of this gene among vertebrates. In addition, we cloned the cDNA encoding a putative 26RFa receptor, G protein-coupled receptor 103 (GPR103) in the zebra finch brain. GPR103 cDNA encoded a 432 amino acid protein that has seven transmembrane domains. In situ hybridisation analysis in the brain showed that the expression of 26RFa mRNA is confined to the anterior-medial hypothalamic area, ventromedial nucleus of the hypothalamus and the lateral hypothalamic area, the brain regions that are involved in the regulation of feeding behaviour, whereas GPR103 mRNA is distributed throughout the brain in addition to the hypothalamic nuclei. When administered centrally in free-feeding male zebra finches, 26RFa increased food intake 24 h after injection without body mass change. Diencephalic GPR103 mRNA expression was up-regulated by fasting for 10 h. Our data suggest that the hypothalamic 26RFa-its receptor system plays an important role in the central control of food intake and energy homeostasis in the zebra finch.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/metabolismo , Pinzones/anatomía & histología , Pinzones/metabolismo , Neuropéptidos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Pollos , Ingestión de Alimentos , Metabolismo Energético , Conducta Alimentaria/fisiología , Femenino , Privación de Alimentos , Perfilación de la Expresión Génica , Homeostasis , Humanos , Masculino , Datos de Secuencia Molecular , Neuropéptidos/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Alineación de Secuencia
10.
Pharmacol Biochem Behav ; 99(3): 500-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21586302

RESUMEN

The purpose of this study was to investigate the effects of acute and chronic administration of anabolic-androgenic steroids (AAS) on nociception and morphine antinociception in acute pain models, as well as on chronic inflammatory nociception. In Experiment 1, adult, gonadally intact male rats were injected s.c. for 28 days with either 5 mg/kg testosterone (T), dihydrotestosterone (DHT), stanozolol (STAN), or safflower oil vehicle (N=12-25/group). On day 28, rats in each group were tested on acute thermal and mechanical nociceptive assays, before and after morphine treatment. In Experiment 2, rats in each group (N=8-10/group) were injected with mineral oil or complete Freund's adjuvant (CFA) into one hindpaw after 28 days of AAS treatment, and then tested for thermal hyperalgesia, mechanical allodynia, inflammation and locomotor suppression intermittently for 28 days. Experiment 3 replicated nociceptive measurements in Experiments 1 and 2, but with a single AAS or vehicle injection occurring 3h prior to testing (N=10-12/group). While chronic AAS administration tended to decrease body weight gain and alter reproductive organ weights in the expected manner, it did not significantly alter acute nociception nor attenuate the development of various chronic pain indices after CFA administration. Morphine antinociceptive potency was significantly decreased by chronic DHT on the hot plate test only. Acute AAS administration also did not significantly alter acute or chronic nociception, or morphine antinociceptive potency. Comparisons between acute and chronic AAS administration suggest that steroid tolerance did not occur in rats treated with AAS chronically. Taken together, these data do not support the hypothesis that AAS exposure alters nociception or morphine antinociception in gonadally intact males.


Asunto(s)
Analgésicos Opioides/farmacología , Dihidrotestosterona/farmacología , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Estanozolol/farmacología , Testosterona/farmacología , Animales , Masculino , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
11.
J Neuroendocrinol ; 23(1): 39-51, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083774

RESUMEN

Among the RFamide peptide family, the LPXRFamide peptide (LPXRFa) group regulates the release of various pituitary hormones and, recently, LPXRFa genes were found to be regulated by photoperiod via melatonin. As a first step towards investigating the role of LPXRFa on reproductive function in grass puffer (Takifugu niphobles), which spawns in semilunar cycles, genes encoding LPXRFa and its receptor (LPXRFa-R) were cloned, and seasonal, diurnal and circadian changes in their absolute amounts of mRNAs in the brain and pituitary were examined by quantitative real-time polymerase chain reaction. The grass puffer LPXRFa precursor contains two putative RFamide peptides and one possible RYamide peptide. LPXRFa and LPXRFa-R genes were extensively expressed in the diencephalon and pituitary. The expression levels of both genes were significantly elevated during the spawning periods in both sexes in the brain and pituitary, although they were low in the spawning fish just after releasing eggs and sperm. The treatment of primary pituitary cultures with goldfish LPXRFa increased the amounts of follicle-stimulating hormone ß- and luteinising hormone ß-subunit mRNAs. In the diencephalon, LPXRFa and LPXRFa-R genes showed synchronised diurnal and circadian variations with one peak at zeitgeber time 3 and circadian time 15, respectively. The correlated expression patterns of LPXRFa and LPXRFa-R genes in the diencephalon and pituitary and the possible stimulatory effects of LPXRFa on gonadotrophin subunit gene expression suggest the functional significance of the LPXRFa and LPXRFa-R system in the regulation of lunar-synchronised spawning of grass puffer.


Asunto(s)
Ritmo Circadiano/fisiología , Neuropéptidos/genética , Receptores de Péptidos/genética , Estaciones del Año , Conducta Sexual Animal , Tetraodontiformes/fisiología , Animales , Secuencia de Bases , Ritmo Circadiano/genética , Clonación Molecular , Cartilla de ADN , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
12.
Pharmacol Biochem Behav ; 96(4): 402-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20600244

RESUMEN

This study was undertaken to determine whether depression-like behavior can be observed in gonadally intact females that have experienced normal pregnancy. When tested on the forced swim test (FST) on postpartum days 1-7, previously pregnant rats spent slightly more time immobile, significantly less time swimming and diving, and defecated more than virgin controls. Subchronic treatment with nomifensine (DA reuptake inhibitor, 2.5mg/kg) but not sertraline (serotonin reuptake inhibitor, 10mg/kg) or desipramine (norepinephrine reuptake inhibitor, 10mg/kg) significantly decreased immobility on postpartum day 2. In rats pre-exposed to the FST in mid-pregnancy, neither subchronic nor chronic treatment with desipramine or sertraline decreased immobility on postpartum day 2; in contrast, chronic desipramine significantly decreased immobility in virgin controls. These results indicate that postpartum female rats, compared to virgin controls, show a reduction in some "active coping behaviors" but no significant increase in immobility when tested during the early postpartum period, unlike ovariectomized females that have undergone hormone-simulated pregnancy (HSP). Additionally, immobility that is increased by FST pre-exposure is not readily prevented by treatment with standard antidepressant medications in postpartum females. Depression-like behaviors previously observed in females that have undergone HSP may result from the more dramatic changes in estradiol, prolactin or corticosterone that occur during the early "postpartum" period, compared to the more subtle changes in these hormones that occur in actual postpartum females.


Asunto(s)
Conducta Animal , Preñez/fisiología , Estrés Fisiológico , Natación , Animales , Antidepresivos/administración & dosificación , Desipramina/administración & dosificación , Femenino , Inhibidores de la Captación de Neurotransmisores/administración & dosificación , Nomifensina/administración & dosificación , Ovariectomía , Embarazo , Ratas , Ratas Sprague-Dawley , Sertralina/administración & dosificación
13.
J Neuroendocrinol ; 22(7): 692-700, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20646173

RESUMEN

To maximise reproductive success, organisms restrict breeding to optimal times of the day or year, when internal physiology and external environmental conditions are suitable for the survival of both parent and offspring. To appropriately coordinate reproductive activity, internal and external standing is communicated to the hypothalamic-pituitary-gonadal axis via a coordinated balance of stimulatory and inhibitory neurochemical systems. The cumulative balance of these mediators ultimately drives the pattern of gonadotrophin-releasing hormone secretion, a neurohormone that stimulates pituitary gonadotrophin secretion. Until 2000, a complementary inhibitor of pituitary gonadotrophin secretion had not been identified. At this time, a novel, avian hypothalamic peptide capable of inhibiting gonadotrophin secretion in cultured quail pituitary cells was uncovered and named gonadotrophin-inhibitory hormone (GnIH). Subsequently, the presence and functional role for the mammalian orthologue of GnIH, RFamide-related peptide, (RFRP-3), was examined, confirming a conserved role for this peptide across several rodent species. To date, a similar distribution and functional role for RFRP-3 have been observed across all mammals investigated, including humans. This overview summarises the role that RFRP-3 plays in mammals and considers the implications and opportunities for further study with respect to reproductive physiology and the neural control of sexual behaviour and motivation.


Asunto(s)
Conducta Animal/fisiología , Conducta/fisiología , Neuropéptidos/metabolismo , Reproducción/fisiología , Animales , Gónadas/metabolismo , Humanos , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Estaciones del Año , Conducta Sexual , Conducta Sexual Animal
14.
J Neuroendocrinol ; 22(7): 716-27, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20456604

RESUMEN

Gonadotrophin-releasing hormone (GnRH) is the primary hypothalamic factor responsible for the control of gonadotrophin secretion in vertebrates. However, within the last decade, two other hypothalamic neuropeptides have been found to play key roles in the control of reproductive functions: gonadotrophin-inhibitory hormone (GnIH) and kisspeptin. In 2000, we discovered GnIH in the quail hypothalamus. GnIH inhibits gonadotrophin synthesis and release in birds through actions on GnRH neurones and gonadotrophs, mediated via GPR147. Subsequently, GnIH orthologues were identified in other vertebrate species from fish to humans. As in birds, mammalian and fish GnIH orthologues inhibit gonadotrophin release, indicating a conserved role for this neuropeptide in the control of the hypothalamic-pituitary-gonadal axis across species. Subsequent to the discovery of GnIH, kisspeptin, encoded by the KiSS-1 gene, was discovered in mammals. By contrast to GnIH, kisspeptin has a direct stimulatory effect on GnRH neurones via GPR54. GPR54 is also expressed in pituitary cells, but whether gonadotrophs are targets for kisspeptin remains unresolved. The KiSS-1 gene is also highly conserved and has been identified in mammals, amphibians and fish. We have recently found a second isoform of KiSS-1, designated KiSS-2, in several vertebrates, but not birds, rodents or primates. In this review, we highlight the discovery, mechanisms of action, and functional significance of these two chief regulators of the reproductive axis.


Asunto(s)
Glicoproteínas/metabolismo , Gonadotropinas/antagonistas & inhibidores , Neuropéptidos/metabolismo , Reproducción/fisiología , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Evolución Biológica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/genética , Gonadotropinas/metabolismo , Humanos , Datos de Secuencia Molecular , Neuronas/metabolismo , Neuropéptidos/clasificación , Neuropéptidos/genética , Filogenia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
15.
J Neuroendocrinol ; 21(4): 322-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19210294

RESUMEN

The optic gland, which is analogous to the anterior pituitary in the context of gonadal maturation, is found on the upper posterior edge of the optic tract of the octopus Octopus vulgaris. In mature octopus, the optic glands enlarge and secrete a gonadotrophic hormone. A peptide with structural features similar to that of vertebrate gonadotrophin-releasing hormone (GnRH) was isolated from the brain of octopus and was named oct-GnRH. Oct-GnRH showed luteinising hormone-releasing activity in the anterior pituitary cells of the Japanese quail Coturnix coturnix. Oct-GnRH immunoreactive signals were observed in the glandular cells of the mature optic gland. Oct-GnRH stimulated the synthesis and release of sex steroids from the ovary and testis, and elicited contractions of the oviduct. Oct-GnRH receptor was expressed in the gonads and accessory organs, such as the oviduct and oviducal gland. These results suggest that oct-GnRH induces the gonadal maturation and oviposition by regulating sex steroidogenesis and a series of egg-laying behaviours via the oct-GnRH receptor. The distribution and expression of oct-GnRH in the central and peripheral nervous systems suggest that oct-GnRH acts as a multifunctional modulatory factor in feeding, memory processing, sensory, movement and autonomic functions.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Octopodiformes/fisiología , Animales , Encéfalo/metabolismo , Conducta Alimentaria/fisiología , Hormona Liberadora de Gonadotropina/genética , Gónadas/metabolismo , Corazón/fisiología , Memoria/fisiología , Actividad Motora/fisiología , Reproducción/fisiología , Conducta Sexual Animal/fisiología
16.
J Neuroendocrinol ; 21(4): 276-81, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19210295

RESUMEN

Gonadotrophin-inhibitory hormone (GnIH) was discovered 8 years ago in birds. Its identification raised the possibility that gonadotrophin-releasing hormone (GnRH) is not the sole hypothalamic neuropeptide that directly influences pituitary gonadotrophin release. Initial studies on GnIH focused on the avian anterior pituitary as comprising the only physiological target of GnIH. There are now several lines of evidence indicating that GnIH directly inhibits pituitary gonadotrophin synthesis and release in birds and mammals. Histological studies on projections from hypothalamic GnIH neurones subsequently implied direct actions of GnIH within the brain and in the periphery. In addition to actions on the pars distalis via the median eminence, GnIH axons and terminals are present in multiple brain areas in birds, and the GnIH receptor is expressed on GnRH-I and -II neurones. Furthermore, we have demonstrated the presence of GnIH and its receptor in avian and mammalian gonads. Thus, GnIH can act directly at multiple levels: within the brain, on the pituitary and in the gonads. In sum, our data indicate that GnIH and its related peptides are important modulators of reproductive function at the level of the GnRH neurone, the gonadotroph and the gonads. Here, we provide an overview of the known levels of GnIH action in birds and mammals. In addition, environmental and physiological factors that are involved in GnIH regulation are reviewed.


Asunto(s)
Encéfalo/metabolismo , Gonadotropinas/metabolismo , Gónadas/crecimiento & desarrollo , Hormonas Hipotalámicas/metabolismo , Hipófisis/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Humanos , Hipotálamo/metabolismo , Neuronas/metabolismo , Periodicidad , Reproducción/fisiología
17.
J Neuroendocrinol ; 21(4): 271-5, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19207818

RESUMEN

In vertebrates, the neuropeptide control of gonadotrophin secretion is primarily through the stimulatory action of the hypothalamic decapeptide, gonadotrophin-releasing hormone (GnRH). Gonadal sex steroids and inhibin inhibit gonadotrophin secretion via feedback from the gonads, but a hypothalamic neuropeptide inhibiting gonadotrophin secretion was, until recently, unknown in vertebrates. In 2000, we discovered a novel hypothalamic dodecapeptide that directly inhibits gonadotrophin release in quail and termed it gonadotrophin-inhibitory hormone (GnIH). GnIH acts on the pituitary and GnRH neurones in the hypothalamus via a novel G-protein-coupled receptor for GnIH to inhibit gonadal development and maintenance by decreasing gonadotrophin release and synthesis. The pineal hormone melatonin is a key factor controlling GnIH neural function. GnIH occurs in the hypothalamus of several avian species and is considered to be a new key neurohormone inhibiting avian reproduction. Thus, the discovery of GnIH provides novel directions to investigate neuropeptide regulation of reproduction. This review summarises the discovery, progress and prospects of GnIH, a new key neurohormone controlling reproduction.


Asunto(s)
Proteínas Aviares/aislamiento & purificación , Proteínas Aviares/metabolismo , Gonadotropinas/metabolismo , Hormonas Hipotalámicas/aislamiento & purificación , Hormonas Hipotalámicas/metabolismo , Animales , Aves , Coturnix , Gónadas/crecimiento & desarrollo , Hipotálamo/metabolismo , Melatonina/metabolismo , Neuronas/metabolismo , Hipófisis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reproducción/fisiología
18.
Eur J Neurosci ; 25(8): 2523-35, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17445248

RESUMEN

The lateral prefrontal cortex (LPFC) is important for working memory (WM) task performance. Neuropsychological and neurophysiological studies in monkeys suggest that the lateral prefrontal cortex is functionally segregated based on the working memory domain (spatial vs. non-spatial). However, this is not supported by most human neuroimaging studies, and the discrepancy might be due to differences in methods and/or species (monkey neuropsychology/physiology vs. human neuroimaging). We used positron emission topography to examine the functional segregation of the lateral prefrontal cortex of Japanese macaques (Macaca fuscata) that showed near 100% accuracy on spatial and non-spatial working memory tasks. Compared with activity during the non-working memory control tasks, the dorsolateral prefrontal cortex (DLPFC) was more active during the non-spatial, but not during the spatial, working memory task, although a muscimol microinjection into the dorsolateral prefrontal cortex significantly impaired the performance of both working memory tasks. A direct comparison of the brain activity between the two working memory tasks revealed no differences within the lateral prefrontal cortex, whereas the premotor area was more active during the spatial working memory task. Comparing the delay-specific activity, which did not include task-associated stimulus/response-related activity, revealed more spatial working memory-related activity in the posterior parietal and premotor areas, and more non-spatial working memory-related activity in the dorsolateral prefrontal cortex and hippocampus. These results suggest that working memory in the monkey brain is segregated based on domain, not within the lateral prefrontal cortex but rather between the posterior parietal-premotor areas and the dorsolateral prefrontal-hippocampus areas.


Asunto(s)
Corteza Cerebral/fisiología , Memoria/fisiología , Animales , Conducta Animal/fisiología , Corteza Cerebral/anatomía & histología , Agonistas del GABA/metabolismo , Humanos , Macaca , Masculino , Muscimol/metabolismo , Pruebas Neuropsicológicas , Tomografía de Emisión de Positrones
19.
J Int Med Res ; 33(6): 654-60, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16372583

RESUMEN

The deposition of amyloid beta (Abeta) protein is a neuropathological change that characterizes Alzheimer's disease. Animals with the osteopetrosis (op/op) mutation suffer from a general skeletal sclerosis, a significantly reduced number of macrophages and osteoclasts in various tissues, and have no systemic macrophage colony stimulating factor (M-CSF). This study examined the effect that M-CSF injections had on Abeta deposition and microglial cell distribution in the brains of normal and op/op mice. Abeta-positive plaques were detected in the cerebral cortex of op/op mice, but not in normal mice. M-CSF reduced the numbers of Abeta-positive plaques in op/op mice. The microglial cell population was reduced in op/op mice compared with normal mice, and M-CSF increased the numbers to 65.8% of that observed in normal mice. Our results suggest that a clearer understanding of the role that microglial cells play in Abeta deposition may help determine the mechanisms involved in the pathogenesis of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Microglía/patología , Osteopetrosis/patología , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Femenino , Inyecciones , Masculino , Ratones , Ratones Mutantes , Microglía/efectos de los fármacos , Microglía/metabolismo , Osteopetrosis/metabolismo , Valores de Referencia
20.
Phys Rev Lett ; 94(20): 207003, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-16090275

RESUMEN

We report a resonant inelastic x-ray scattering (RIXS) study of charge excitations in the electron-doped high-T(c) superconductor Nd1.85 Ce0.15 CuO4. The intraband and interband excitations across the Fermi energy are separated for the first time by tuning the experimental conditions properly to measure charge excitations at low energy. A dispersion relation with q-dependent width emerges clearly in the intraband excitation, while the intensity of the interband excitation is concentrated around 2 eV near the zone center. The experimental results are consistent with theoretical calculation of the RIXS spectra based on the Hubbard model.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...