Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biol Reprod ; 105(4): 1056-1067, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34037695

RESUMEN

Mechanisms in the brain controlling secretion of gonadotropin hormones in pigs, particularly luteinizing hormone (LH), are poorly understood. Kisspeptin is a potent LH stimulant that is essential for fertility in many species, including pigs. Neurokinin B (NKB) acting through neurokinin 3 receptor (NK3R) is involved in kisspeptin-stimulated LH release, but organization of NKB and NK3R within the porcine hypothalamus is unknown. Hypothalamic tissue from ovariectomized (OVX) gilts was used to determine the distribution of immunoreactive kisspeptin, NKB, and NK3R cells in the arcuate nucleus (ARC). Almost all kisspeptin neurons coexpressed NKB in the porcine ARC. Immunostaining for NK3R was distributed throughout the preoptic area (POA) and in several hypothalamic areas including the periventricular and retrochiasmatic areas but was not detected within the ARC. There was no colocalization of NK3R with gonadotropin-releasing hormone (GnRH), but NK3R-positive fibers in the POA were in close apposition to GnRH neurons. Treating OVX gilts with the progestin altrenogest decreased LH pulse frequency and reduced mean circulating concentrations of LH compared with OVX control gilts (P < 0.01), but the number of kisspeptin and NKB cells in the ARC did not differ between treatments. The neuroanatomical arrangement of kisspeptin, NKB, and NK3R within the porcine hypothalamus confirms they are positioned to stimulate GnRH and LH secretion in gilts, though differences with other species exist. Altrenogest suppression of LH secretion in the OVX gilt does not appear to involve decreased peptide expression of kisspeptin or NKB.


Asunto(s)
Hipotálamo/metabolismo , Kisspeptinas/genética , Neuroquinina B/genética , Progestinas/farmacología , Receptores de Neuroquinina-3/genética , Sus scrofa/genética , Acetato de Trembolona/análogos & derivados , Animales , Femenino , Perfilación de la Expresión Génica/veterinaria , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Receptores de Neuroquinina-3/metabolismo , Sus scrofa/metabolismo , Acetato de Trembolona/farmacología
2.
Gen Comp Endocrinol ; 281: 126-136, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31163181

RESUMEN

To ascertain the significance of the Neurokinin B/Tachykinin 3 receptor (NKB/NK3) system in goldfish reproduction, two cDNAs encoding tachykinin 3 receptors, namely tacr3a and tacr3b, were cloned. Subsequent studies revealed that the downstream signalling of both Tac3rs can be activated by different NKB peptides, suggesting that the cloned receptors are biologically functional in goldfish. RT-PCR analysis showed that tacr3s are widely expressed in brain regions. During the gonadal development, tacr3a and tacr3b exhibited different expression patterns in the hypothalamus and pituitary. The actions of NKB peptides on reproductive axis was further investigated in vivo. Intraperitoneal injections of NKB peptides significantly reduced the expression of kiss2 and gonadotropin releasing hormone 3 (gnrh3) in the hypothalamus, and the expression of luteinizing hormone beta subunit (lhb) and follicle stimulating hormone beta subunit (fshb) in the pituitary in sexually immature goldfish. Taken together, our findings revealed that NKB/NK3 system plays a negative role in the reproductive axis of immature goldfish.


Asunto(s)
Carpa Dorada/fisiología , Neuroquinina B/metabolismo , Receptores de Neuroquinina-3/metabolismo , Reproducción/fisiología , Maduración Sexual , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Células HEK293 , Humanos , Hipotálamo/metabolismo , Masculino , Filogenia , Hipófisis/metabolismo , ARN Mensajero/metabolismo , Receptores de Neuroquinina-3/química , Receptores de Neuroquinina-3/genética
3.
Gynecol Endocrinol ; 34(5): 437-441, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29187003

RESUMEN

The effects of androgens on gonadotropin-releasing hormone (GnRH) secretion in females have not been fully established. To clarify the direct effects of androgens on hypothalamic reproductive factors, we evaluated the effects of chronic testosterone administration on hypothalamic GnRH regulatory factors in ovariectomized (OVX) female rats. Both testosterone and estradiol reduced the serum luteinizing hormone levels of OVX female rats, indicating that, as has been found for estrogen, testosterone suppresses GnRH secretion via negative feedback. Similarly, the administration of testosterone or estradiol suppressed the hypothalamic mRNA levels of kisspeptin and neurokinin B, both of which are positive regulators of GnRH, whereas it did not affect the hypothalamic mRNA levels of the kisspeptin receptor or neurokinin-3 receptor. On the contrary, the administration of testosterone, but not estradiol, suppressed the hypothalamic mRNA expression of prodynorphin, which is a negative regulator of GnRH. The administration of testosterone did not alter the rats' serum estradiol levels, indicating that testosterone's effects on hypothalamic factors might be induced by its androgenic activity. These findings suggest that as well as estrogen, androgens have negative feedback effects on GnRH in females and that the underlying mechanisms responsible for these effects are similar, but do not completely correspond, to the mechanisms underlying the effects of estrogen on GnRH.


Asunto(s)
Dinorfinas/metabolismo , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Testosterona/farmacología , Animales , Dinorfinas/genética , Estradiol/farmacología , Femenino , Hipotálamo/metabolismo , Kisspeptinas/genética , Leptina/sangre , Hormona Luteinizante/sangre , Neuroquinina B/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Receptores de Neuroquinina-3/genética , Receptores de Neuroquinina-3/metabolismo
4.
Endocr J ; 58(11): 1003-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21979277

RESUMEN

Kisspeptin, which is the product of the kiss1 gene and its receptor kiss1r, have emerged as the essential gatekeepers of reproduction. The present study used gonadally intact female rats to evaluate fasting-induced suppression of the KiSS-1 system of anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) under normal physiological conditions. Starting on the day of estrous, one group of rats was subjected to 72 h of food deprivation, while the other group of rats was able to continue feeding ad libitum. The length of the estrous cycle was significantly longer in the food-deprived rats as compared to the feeding rats. At the end of the 72-h food deprivation period, all of the food-deprived rats were at the diestrous phase, with their serum concentrations of LH and leptin significantly lower than that observed in the feeding rats. In addition, as compared to the feeding rats, the expression levels of kiss1 mRNA were significantly lower in the food-deprived rats in the posterior hypothalamic block, which contained the ARC, but not in the anterior hypothalamic block, which contain the AVPV. However, both the kiss1r mRNA expression levels in the anterior and posterior hypothalamic blocks and the neurokinin B and neurokinin 3 receptor mRNA expression levels in the posterior hypothalamic block were not significantly different between the feeding and food-deprived rats. Thus, lower kiss1 mRNA levels in the ARC appear to be responsible for the fasting-induced inhibition of gonadotrophin secretion and subsequent prolongation of the estrous cycle.


Asunto(s)
Privación de Alimentos/fisiología , Hipotálamo/fisiología , Kisspeptinas/metabolismo , ARN Mensajero/metabolismo , Animales , Ciclo Estral/fisiología , Femenino , Hipotálamo/metabolismo , Kisspeptinas/genética , Leptina/sangre , Hormona Luteinizante/sangre , Neuroquinina B/genética , Neuroquinina B/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores de Neuroquinina-3/genética , Receptores de Neuroquinina-3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Brain Res ; 1364: 116-28, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20800582

RESUMEN

Loss-of-function mutations in the genes encoding either neurokinin B (NKB) or its receptor, NK3 (NK3R), result in hypogonadotropic hypogonadism, characterized by an absence of pubertal development and low circulating levels of LH and gonadal steroids. These studies implicate NKB and NK3R as essential elements of the human reproductive axis. Studies over the last two decades provide evidence that a group of neurons in the hypothalamic infundibular/arcuate nucleus form an important component of this regulatory circuit. These neurons are steroid-responsive and coexpress NKB, kisspeptin, dynorphin, NK3R, and estrogen receptor α (ERα) in a variety of mammalian species. Compelling evidence in the human indicates these neurons function in the hypothalamic circuitry regulating estrogen negative feedback on gonadotropin-releasing hormone (GnRH) secretion. Moreover, in the rat, they form a bilateral, interconnected network that projects to NK3R-expressing GnRH terminals in the median eminence. This network provides an anatomical framework to explain how coordination among NKB/kisspeptin/dynorphin/NK3R/ERα neurons could mediate feedback information from the gonads to modulate pulsatile GnRH secretion. There is substantial (but indirect) evidence that this network may be part of the neural circuitry known as the "GnRH pulse generator," with NK3R signaling as an important component. This theory provides a compelling explanation for the occurrence of hypogonadotropic hypogonadism in patients with inactivating mutations in the TAC3 or TACR3 genes. Future studies will be needed to determine whether NKB signaling plays a permissive role in the onset of puberty or is part of the driving force initiating the maturation of reproductive function.


Asunto(s)
Hipotálamo/fisiología , Neuroquinina B/fisiología , Reproducción/fisiología , Envejecimiento/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/fisiología , Dinorfinas/fisiología , Estrógenos/fisiología , Femenino , Expresión Génica/genética , Humanos , Kisspeptinas , Macaca mulatta , Masculino , Neuronas/fisiología , Posmenopausia/fisiología , Embarazo , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/genética , Receptores de Neuroquinina-3/fisiología , Caracteres Sexuales , Transducción de Señal/fisiología , Terminología como Asunto , Proteínas Supresoras de Tumor/fisiología
6.
Neuroendocrinology ; 92(2): 81-99, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606386

RESUMEN

Evolutionary survival of a species is largely a function of its reproductive fitness. In mammals, a sparsely populated and widely dispersed network of hypothalamic neurons, the gonadotropin-releasing hormone (GnRH) neurons, serve as the pilot light of reproduction via coordinated secretion of GnRH. Since it first description, human GnRH deficiency has been recognized both clinically and genetically as a heterogeneous disease. A spectrum of different reproductive phenotypes comprised of congenital GnRH deficiency with anosmia (Kallmann syndrome), congenital GnRH deficiency with normal olfaction (normosmic idiopathic hypogonadotropic hypogonadism), and adult-onset hypogonadotropic hypogonadism has been described. In the last two decades, several genes and pathways which govern GnRH ontogeny have been discovered by studying humans with GnRH deficiency. More importantly, detailed study of these patients has highlighted the emerging theme of oligogenicity and genotypic synergism, and also expanded the phenotypic diversity with the documentation of reversal of GnRH deficiency later in adulthood in some patients. The underlying genetic defect has also helped understand the associated nonreproductive phenotypes seen in some of these patients. These insights now provide practicing clinicians with targeted genetic diagnostic strategies and also impact on clinical management.


Asunto(s)
Hormona Liberadora de Gonadotropina/deficiencia , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/crecimiento & desarrollo , Síndrome de Kallmann/genética , Animales , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo , Humanos , Hipogonadismo/genética , Masculino , Ratones , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Trastornos del Olfato/genética , Fenotipo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/metabolismo , Receptores de Neuroquinina-3/genética , Receptores de Neuroquinina-3/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Horm Res Paediatr ; 73(6): 477-81, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20395662

RESUMEN

BACKGROUND/AIMS: TAC3 and TACR3 have recently been shown to be causative genes for an autosomal recessive form of isolated hypogonadotropic hypogonadism (IHH). Here, we report a Japanese female with IHH and compound heterozygous TACR3 mutations and her heterozygous parents, and discuss the primary lesion for IHH and clinical findings. CASE REPORT: This female was identified through mutation analysis of TAC3 and TACR3 in 57 patients with IHH. At 24 years of age, an initial standard GnRH test showed poor gonadotropin response (LH <0.2-0.6 IU/l), whereas the second GnRH test performed after GnRH priming (100 microg i.m. for 5 consecutive days) revealed ameliorated gonadotropin responses (LH 0.3-6.4 IU/l; FSH 2.2-9.6 IU/l). The mother exhibited several features suggestive of mild IHH, whereas the father showed an apparently normal phenotype. RESULTS: She had a paternally derived nonsense mutation at exon 1 (Y145X) and a maternally inherited single nucleotide (G) deletion from the conserved 'GT' splice donor site of intron 1 (IVS1+1delG). CONCLUSIONS: The results suggest hypothalamic dysfunction as the primary cause for IHH in patients with biallelic TACR3 mutations and clinical manifestation in heterozygous females, together with the rarity of TAC3 and TACR3 mutations in patients with IHH.


Asunto(s)
Hipogonadismo/genética , Hipogonadismo/fisiopatología , Hipotálamo/fisiopatología , Mutación/genética , Receptores de Neuroquinina-3/genética , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Fenotipo , Reacción en Cadena de la Polimerasa , Adulto Joven
8.
Neuroscience ; 119(4): 1113-45, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12831868

RESUMEN

As melanin-concentrating hormone (MCH) neurons express the neurokinin 3 receptor (NK3) in the rat diencephalon, their innervation by tachykininergic fibers, the origin of this innervation and the effect of a NK3 agonist on MCH mRNA expression were researched. The obtained results show that the tachykininergic system develops complex relationships with MCH neurons. Overall, MCH cell bodies appeared targeted by both NKB- and SP-inputs. These afferents have multiple hypothalamic and extra-hypothalamic origins, but a local (intra-lateral hypothalamic area) origin from small interneurons was suspected as well. MCH cell bodies do not express NK1, but around 2.7% of the MCH neurons contained SP after colchicine injection. Senktide, a NK3 agonist, produced an increase of the MCH mRNA expression in cultured hypothalamic slices. This effect was reversed by two NK3 antagonists. Tachykinins enhance MCH mRNA expression, and, thus, may modulate the effect of MCH in functions such as feeding and reproductive behaviors in which this peptide has been experimentally involved.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Neuroquinina-3/genética , Sustancia P/análogos & derivados , Taquicininas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Colchicina/metabolismo , Conducta Alimentaria/fisiología , Hipotálamo/citología , Interneuronas/citología , Interneuronas/metabolismo , Masculino , Vías Nerviosas/citología , Neuroquinina B/agonistas , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/metabolismo , Antagonistas del Receptor de Neuroquinina-1 , Neuronas/citología , Técnicas de Cultivo de Órganos , Fragmentos de Péptidos/farmacología , Piperidinas/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/antagonistas & inhibidores , Conducta Sexual/fisiología , Sustancia P/metabolismo , Sustancia P/farmacología
9.
Mol Pharmacol ; 58(3): 552-9, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10953048

RESUMEN

There have been proposals that the tachykinin receptor classification should be extended to include a novel receptor, the "neurokinin-4" receptor (NK-4R), which has a close homology with the human NK-3 receptor (hNK-3R). We compared the pharmacological and molecular biological characteristics of the hNK-3R and NK-4R. Binding experiments, with (125)I-[MePhe(7)]-NKB binding to HEK 293 cell membranes transiently expressing the hNK-3R (HEK 293-hNK-3R) or NK-4R (HEK 293-NK-4R), and functional studies (Ca(2+) mobilization in the same cells) revealed a similar profile of sensitivity to tachykinin agonists and antagonists for both receptors; i.e., in binding studies with the hNK-3R, MePhe(7)-NKB > NKB > senktide >> NKA = Substance P; with the NK-4R, MePhe(7)-NKB > NKB = senktide >> Substance P = NKA; and with antagonists, SB 223412 = SR 142801 > SB 222200 >> SR 48968 >> CP 99994 for both hNK-3R and NK-4R. Thus, the pharmacology of the two receptors was nearly identical. However, attempts to isolate or identify the NK-4R gene by using various molecular biological techniques were unsuccessful. Procedures, including nested polymerase chain reaction studies, that used products with restriction endonuclease sites specific for either hNK-3R or NK-4R, failed to demonstrate the presence of NK-4R in genomic DNA from human, monkey, mouse, rat, hamster, or guinea pig, and in cDNA libraries from human lung, brain, or heart, whereas the hNK-3R was detectable in the latter libraries. In view of the failure to demonstrate the presence of the putative NK-4R it is thought to be premature to extend the current tachykinin receptor classification.


Asunto(s)
Receptores de Neuroquinina-3/metabolismo , Receptores de Taquicininas/metabolismo , Unión Competitiva , Transporte Biológico , Calcio/metabolismo , Células Cultivadas , ADN Complementario/análisis , Humanos , Reacción en Cadena de la Polimerasa , Ensayo de Unión Radioligante , Receptores de Neuroquinina-3/efectos de los fármacos , Receptores de Neuroquinina-3/genética , Receptores de Taquicininas/efectos de los fármacos , Receptores de Taquicininas/genética , Receptores de Taquicininas/aislamiento & purificación , Mapeo Restrictivo , Taquicininas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA