Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
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.
Dev Neurobiol ; 75(1): 12-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25044785

RESUMEN

Members of the tachykinin family have trophic effects on developing neurons. The tachykinin neurokinin 3 receptor (NK3R) appears early in embryonic development; during the peak birthdates of hypothalamic neurons, but its involvement in neural development has not been examined. To address its possible role, immortalized embryonic hypothalamic neurons (CLU209) were treated with CellMask, a plasma membrane stain, or the membranes were imaged in CLU209 cells that were transfected with a pEGFP-NK3R expression vector. Nontransfected cells and transfected cells were then treated with senktide, a NK3R agonist, or Dulbecco's Modified Eagle's Medium (DMEM) and time-lapse confocal images were captured for the following 30 min. Compared to DMEM, senktide treatment led to filopodia initiation from the soma of both nontransfected and transfected CLU209 cells. These filopodia had diameters and lengths of approximately 200 nm and 3 µm, respectively. Pretreatment with an IP3 receptor blocker, 2-aminoethoxydiphenyl borate (2-APB), prevented the senktide-induced growth in filopodia; demonstrating that NK3R-induced outgrowth of filopodia likely involves the release of intracellular calcium. Exposure of transfected CLU209 cells to senktide for 24 h led to further growth of filopodia and processes that extended 10-20 µm. A mathematical model, composed of a linear and population model was developed to account for the dynamics of filopodia growth during a timescale of minutes. The results suggest that the ligand-induced activation of NK3R affects early developmental processes by initiating filopodia formation that are a prerequisite for neuritogenesis.


Asunto(s)
Embrión de Mamíferos/fisiología , Hipotálamo/fisiología , Seudópodos/fisiología , Receptores de Neuroquinina-3/fisiología , Animales , Células Cultivadas , Hipotálamo/embriología , Seudópodos/metabolismo , Ratas , Receptores de Neuroquinina-3/metabolismo
5.
Neuroendocrinology ; 100(2-3): 250-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25428554

RESUMEN

The continuous activation of the kisspeptin receptor by its agonists causes the abrogation of kisspeptin signaling, leading to decreased pulsatile luteinizing hormone (LH) secretion. Employing this phenomenon as a tool for probing kisspeptin action, this study aimed to clarify the role of kisspeptin in gonadotropin-releasing hormone (GnRH) pulse generation in goats. We examined the effects of chronic administration of TAK-683, an investigational kisspeptin analog, on LH secretion, GnRH immunostaining, pituitary responses to exogenous GnRH, and GnRH pulse generator activity, reflected by a characteristic increase in multiple-unit activity (MUA volley). An osmotic pump containing TAK-683 was subcutaneously implanted on day 0. TAK-683 treatment dose-dependently suppressed pulsatile LH secretion on day 1. Higher doses of chronic TAK-683 profoundly suppressed pulsatile LH secretion but had little effect on GnRH immunostaining patterns and pituitary responses to GnRH on day 5. In ovariectomized goats, MUA volleys occurred at approximately every 30 min on day -1. On day 5 of chronic TAK-683 administration, pulsatile LH secretion was markedly suppressed, whereas MUA volleys were similar to those observed on day -1. Male pheromones and senktide (neurokinin B receptor agonist) induced an MUA volley but had no effect on LH secretion during chronic TAK-683 administration. The results indicate that the chronic administration of a kisspeptin analog profoundly suppresses pulsatile LH secretion without affecting GnRH content, pituitary function or GnRH pulse generator activity, and they suggest an indispensable role for kisspeptin signaling in the cascade driving GnRH/LH pulses by the GnRH pulse generator.


Asunto(s)
Relojes Biológicos/efectos de los fármacos , Fármacos del Sistema Nervioso Central/administración & dosificación , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Kisspeptinas/administración & dosificación , Animales , Relojes Biológicos/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Cabras , Hipodermoclisis , Hipotálamo/fisiología , Bombas de Infusión Implantables , Hormona Luteinizante/metabolismo , Masculino , Fragmentos de Péptidos/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/fisiología , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/metabolismo , Sustancia P/análogos & derivados , Sustancia P/farmacología , Testosterona/farmacología
6.
J Neuroendocrinol ; 26(11): 776-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25040132

RESUMEN

Neurokinin B (NKB) is essential for human reproduction and has been shown to stimulate luteinising hormone (LH) secretion in several species, including sheep. Ewes express the neurokinin-3 receptor (NK3R) in the retrochiasmatic area (RCh) and there is one report that placement of senktide, an NK3R agonist, therein stimulates LH secretion that resembles an LH surge in ewes. In the present study, we first confirmed that local administration of senktide to the RCh produced a surge-like increase in LH secretion, and then tested the effects of this agonist in two other areas implicated in the control of LH secretion and where NK3R is found in high abundance: the preoptic area (POA) and arcuate nucleus (ARC). Bilateral microimplants containing senktide induced a dramatic surge-like increase in LH when given in the POA similar to that seen with RCh treatment. By contrast, senktide treatment in the ARC resulted in a much smaller but significant increase in LH concentrations suggestive of an effect on tonic secretion. The possible role of POA and RCh NK3R activation in the LH surge was next tested by treating ewes with SB222200, an NK3R antagonist, in each area during an oestradiol-induced LH surge. SB222200 in the RCh, but not in the POA, reduced the LH surge amplitude by approximately 40% compared to controls, indicating that NK3R activation in the former region is essential for full expression of the pre-ovulatory LH surge. Based on these data, we propose that the actions of NKB in the RCh are an important component of the pre-ovulatory LH surge in ewes.


Asunto(s)
Hipotálamo/efectos de los fármacos , Hormona Luteinizante/sangre , Ovulación/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Neuroquinina-3/agonistas , Sustancia P/análogos & derivados , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Hipotálamo/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovulación/metabolismo , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Quinolinas/farmacología , Receptores de Neuroquinina-3/antagonistas & inhibidores , Receptores de Neuroquinina-3/metabolismo , Ovinos , Sustancia P/farmacología
7.
Front Neuroendocrinol ; 34(3): 211-27, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23872331

RESUMEN

Despite affecting millions of individuals, the etiology of hot flushes remains unknown. Here we review the physiology of hot flushes, CNS pathways regulating heat-dissipation effectors, and effects of estrogen on thermoregulation in animal models. Based on the marked changes in hypothalamic kisspeptin, neurokinin B and dynorphin (KNDy) neurons in postmenopausal women, we hypothesize that KNDy neurons play a role in the mechanism of flushes. In the rat, KNDy neurons project to preoptic thermoregulatory areas that express the neurokinin 3 receptor (NK3R), the primary receptor for NKB. Furthermore, activation of NK3R in the median preoptic nucleus, part of the heat-defense pathway, reduces body temperature. Finally, ablation of KNDy neurons reduces cutaneous vasodilatation and partially blocks the effects of estrogen on thermoregulation. These data suggest that arcuate KNDy neurons relay estrogen signals to preoptic structures regulating heat-dissipation effectors, supporting the hypothesis that KNDy neurons participate in the generation of flushes.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Dinorfinas/fisiología , Sofocos/fisiopatología , Hipotálamo/metabolismo , Kisspeptinas/fisiología , Neuroquinina B/fisiología , Animales , Regulación de la Temperatura Corporal/efectos de los fármacos , Estradiol/farmacología , Ciclo Estral/efectos de los fármacos , Femenino , Humanos , Hormona Luteinizante/metabolismo , Modelos Biológicos , Neuronas/fisiología , Ovariectomía , Posmenopausia/fisiología , Área Preóptica/metabolismo , Ratas , Receptores de Neuroquinina-3/metabolismo , Transducción de Señal , Piel/irrigación sanguínea , Cola (estructura animal)/irrigación sanguínea , Vasodilatación
8.
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
9.
Reprod Toxicol ; 31(4): 440-6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21185367

RESUMEN

Mechanism mediating the testicular toxicity induced by CS-003, a triple neurokinin receptor antagonist, was investigated in male dogs. Daily CS-003 administrations showed testicular toxicity, such as a decrease in the sperm number, motility and prostate weight; and an increase in sperm abnormality, accompanying histopathological changes in the testis, epididymis and prostate. A single CS-003 administration suppressed plasma testosterone and LH levels in intact and castrated males. The suppressed LH release was restored by GnRH agonist injection, suggesting that pituitary sensitivity to GnRH is not impaired by CS-003. Treatment with SB223412, a neurokinin 3 receptor antagonist, caused a similar effect to CS-003, such as toxicity in the testis, prostate and epididymis and decreased plasma level of LH and testosterone. In conclusion, CS-003-induced testicular toxicity is caused by the inhibition of neurokinin B/neurokinin 3 receptor signaling probably at the hypothalamic level in male dogs.


Asunto(s)
Óxidos S-Cíclicos/toxicidad , Hipotálamo/efectos de los fármacos , Morfolinas/toxicidad , Receptores de Taquicininas/antagonistas & inhibidores , Espermatozoides/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Perros , Hormona Folículo Estimulante/sangre , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Masculino , Neuroquinina B/metabolismo , Orquiectomía , Tamaño de los Órganos/efectos de los fármacos , Próstata/efectos de los fármacos , Próstata/patología , Quinolinas/toxicidad , Receptores de Neuroquinina-3/antagonistas & inhibidores , Receptores de Neuroquinina-3/metabolismo , Receptores de Taquicininas/metabolismo , Recuento de Espermatozoides , Motilidad Espermática/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatozoides/metabolismo , Espermatozoides/patología , Testículo/metabolismo , Testículo/patología , Testosterona/sangre , Factores de Tiempo
10.
PLoS One ; 5(11): e15471, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21103352

RESUMEN

Sub-populations of neurons producing melanin-concentrating hormone (MCH) are characterized by distinct projection patterns, birthdates and CART/NK3 expression in rat. Evidence for such sub-populations has not been reported in other species. However, given that genetically engineered mouse lines are now commonly used as experimental models, a better characterization of the anatomy and morphofunctionnal organization of MCH system in this species is then necessary. Combining multiple immunohistochemistry experiments with in situ hybridization, tract tracing or BrdU injections, evidence supporting the hypothesis that rat and mouse MCH systems are not identical was obtained: sub-populations of MCH neurons also exist in mouse, but their relative abundance is different. Furthermore, divergences in the distribution of MCH axons were observed, in particular in the ventromedial hypothalamus. These differences suggest that rat and mouse MCH neurons are differentially involved in anatomical networks that control feeding and the sleep/wake cycle.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Neuroquinina-3/metabolismo , Animales , Axones/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormonas Hipotalámicas/genética , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Melaninas/genética , Ratones , Hormonas Hipofisarias/genética , Ratas , Ratas Long-Evans , Especificidad de la Especie , Factores de Tiempo
11.
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
12.
J Neuroendocrinol ; 22(1): 1-12, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19912479

RESUMEN

Recent evidence has implicated neurokinin B (NKB) in the complex neuronal network mediating the effects of gonadal steroids on the regulation of gonadotrophin-releasing hormone (GnRH) secretion. Because the neurokinin 3 receptor (NK3R) is considered to mediate the effects of NKB at the cellular level, we determined the distribution of immunoreactive NK3R in the septal region, preoptic area (POA) and hypothalamus of the ewe. NK3R cells and/or fibres were found in areas including the bed nucleus of the stria terminalis, POA, anterior hypothalamic and perifornical areas, dopaminergic A15 region, dorsomedial and lateral hypothalamus, arcuate nucleus (ARC) and the ventral premammillary nucleus. We also used dual-label immunocytochemistry to determine whether a neuroanatomical basis for direct modulation of GnRH neurones by NKB was evident. No GnRH neurones at any rostral-caudal level were observed to contain NK3R immunoreactivity, although GnRH neurones and fibres were in proximity to NK3R-containing fibres. Because NKB fibres formed close contacts with NKB neurones in the ARC, we determined whether these NKB neurones also contained immunoreactive NK3R. In luteal-phase ewes, 64% +/- 11 of NKB neurones colocalised NK3R. In summary, NK3R is distributed in areas of the sheep POA and hypothalamus known to be involved in the control of reproductive neuroendocrine function. Colocalisation of NK3R in NKB neurones of the ARC suggests a potential mechanism for the autoregulation of this subpopulation; however, the lack of NK3R in GnRH neurones suggests that the actions of NKB on GnRH neurosecretory activity in the ewe are mediated indirectly via other neurones and/or neuropeptides.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Receptores de Neuroquinina-3/metabolismo , Núcleos Septales/metabolismo , Animales , Recuento de Células , Femenino , Técnica del Anticuerpo Fluorescente , Microscopía Confocal , Red Nerviosa/metabolismo , Ovinos
13.
Am J Physiol Regul Integr Comp Physiol ; 293(2): R931-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17522129

RESUMEN

Activation of the neurokinin 3 receptor (NK3R) by a receptor agonist, hypotension, and hyperosmolarity results in the internalization of NK3R expressed by magnocellular neurons and the release of vasopressin (VP) and oxytocin (OT) into the circulation. The contribution of NK3R activation to the release of VP and OT in response to hyperosmolarity and hypotension was evaluated by measuring the release of both hormones following pretreatment with a selective NK3R antagonist, SB-222200. Freely behaving male rats were given an intraventricular injection of either 0.15 M NaCl or 250, 500, or 1,000 pmol SB-222200, and then were administered an intravenous infusion of 2 M NaCl or 0.15 M NaCl (experiment 1), or a bolus intra injection of 0.15 M NaCl or hydralazine (HDZ), a hypotension-inducing drug (experiment 2). Blood samples were taken from indwelling arterial catheters at various time points for 1-2 h, both before and after treatments. Plasma VP and OT levels were determined by ELISA. Blockade of NK3R did not affect the baseline levels of either hormone. In contrast, pretreatment with SB-222200 significantly reduced ( approximately 60%) or abolished the release of VP and OT, respectively, to 2 M NaCl infusion. HDZ-induced VP and OT release was eliminated by pretreatment with 500 pmol SB-222200. Therefore, NK3R activation contributes significantly to the systemic release of both VP and OT in response to osmotic and hypotensive challenges.


Asunto(s)
Hipotensión/fisiopatología , Oxitocina/sangre , Receptores de Neuroquinina-3/metabolismo , Vasopresinas/sangre , Equilibrio Hidroelectrolítico/fisiología , Animales , Hidralazina/farmacología , Hipotensión/inducido químicamente , Hipotensión/metabolismo , Hipotálamo/fisiología , Infusiones Intravenosas , Inyecciones Intraventriculares , Masculino , Presión Osmótica , Quinolinas/farmacología , Ratas , Receptores de Neuroquinina-3/antagonistas & inhibidores , Cloruro de Sodio/farmacología , Vasodilatadores/farmacología , Equilibrio Hidroelectrolítico/efectos de los fármacos
14.
Regul Pept ; 125(1-3): 125-33, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15582723

RESUMEN

Nonneuronal cell sources of tachykinins, such as substance P (SP) and neurokinin B (NKB), have been demonstrated in leukocytes, endothelial cells and endocrine cells, and may play a role in corpus luteum (CL) development. For this reason, we analyzed mRNA presence for the two tachykinin precursors together with the neurokinin-1 receptor and the neurokinin-3 receptor (NK-1R and NK-3R, preferred by SP and NKB, respectively) in bovine CL at various stages in the luteal phase. Using the RT-PCR technique, we detected coexpression for the preprotachykinin A gene (PPT-A), which encodes SP and neurokinin A (NKA), and the preprotachykinin B gene (PPT-B) for NKB in the CL at the development, secretion and regression stages. Coexpression was also noted for NK-1R and NK-3R gene transcripts. Cultures of endothelial cells (ECs) derived from bovine CL expressed NK-1R and NK-3R mRNA, as did ovarian macrophages. Agonist treatment induced a stronger intracellular calcium ([Ca2+]i) increase after activation of NK-1R compared to NK-3R, a result that we verified by calcium imaging. This is the first evidence for functional tachykinin receptor activity in luteal ECs and ovarian macrophages from bovine CL.


Asunto(s)
Cuerpo Lúteo/metabolismo , Endotelio/metabolismo , Macrófagos/citología , Ovario/metabolismo , Precursores de Proteínas/biosíntesis , ARN Mensajero/metabolismo , Receptores de Taquicininas/metabolismo , Taquicininas/biosíntesis , Animales , Calcio/metabolismo , Bovinos , Clonación Molecular , Cartilla de ADN/química , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Femenino , Humanos , Macrófagos/metabolismo , ARN/metabolismo , Receptores de Neuroquinina-3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Venas Umbilicales/citología
15.
Neuroscience ; 112(3): 639-45, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12074905

RESUMEN

Although the pathophysiology of primary dystonias is currently unknown, it is thought to involve changes in the basal ganglia-thalamus-cortex circuit, particularly activity imbalances between direct and indirect striatal pathways. Substance P, a member of the tachykinin family of neuropeptides, is a major component in the direct pathway from striatum to basal ganglia output nuclei. In the present study quantitative autoradiography was used to examine changes in neurokinin-1 (NK-1) and neurokinin-3 (NK-3) receptors in mutant dystonic hamsters (dt(sz)), a well characterized model of paroxysmal dystonia. NK-1 receptors were labeled in 10 dystonic brains and 10 age-matched controls with 3 nM [(3)H]-[Sar(9), Met(O(2))(11)]-SP. NK-3 binding sites were labeled in adjacent sections with 2.5 nM [(3)H]senktide. NK-1 binding was found to be unaltered in 27 brain areas examined. In contrast, NK-3 binding was significantly reduced in layers 4 and 5 of the prefrontal (-46%), anterior cingulate (-42%) and parietal (-45%) cortices, ventromedial thalamus (-42%) and substantia nigra pars compacta (-36%) in dystonic brains compared to controls. The latter effects may be particularly relevant in view of evidence that activation of NK-3 receptors on dopaminergic neurons in the substantia nigra pars compacta can increase nigrostriatal dopaminergic activity. Since previous studies indicated that a reduced basal ganglia output in mutant hamsters is based on an overactivity of the direct pathway which also innervates substantia nigra pars compacta neurons, the decreased NK-3 binding could be related to a receptor down-regulation. The present finding of decreased NK-3 receptor density in the substantia nigra pars compacta, thalamic and cortical areas substantiates the hypothesis that disturbances of the basal ganglia-thalamus-cortex circuit play a critical role in the pathogenesis of paroxysmal dystonia.


Asunto(s)
Encéfalo/metabolismo , Distonía/metabolismo , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuroquinina-3/metabolismo , Animales , Ganglios Basales/metabolismo , Corteza Cerebral/metabolismo , Cricetinae , Distonía/genética , Femenino , Masculino , Valores de Referencia , Tálamo/metabolismo
16.
Eur J Neurosci ; 13(9): 1733-44, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11359525

RESUMEN

The ontogeny of rat diencephalic melanin-concentrating hormone (MCH) neurons has been analysed, using the bromodeoxyuridine method to determine the period of birth of these neurons, and using in situ hybridization and immunohistochemistry to study their chemical differentiation. The spatiotemporal pattern of MCH neuron generation is complex, although it is broadly lateromedial with a peak between embryonic days (E) 12 and E13. The first expression of the MCH gene has been detected on E13 in neurons in the presumptive lateral hypothalamic area. But the adult-like pattern was observed from E18. Medial-most MCH neurons express the peptide CART (cocaine-amphetamine-regulated transcript) from E18, and the receptor neurokinin 3 (NK3) from between postnatal day (P) 0 and P5. These results are discussed and compared with data from the literature to better understand the organization of the 'MCH-containing area'.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hormonas Hipotalámicas/metabolismo , Hipotálamo/embriología , Melaninas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Células Madre/metabolismo , Animales , Animales Recién Nacidos , Bromodesoxiuridina/farmacocinética , Embrión de Mamíferos , Hormonas Hipotalámicas/genética , Hipotálamo/citología , Hipotálamo/crecimiento & desarrollo , Inmunohistoquímica , Melaninas/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Hormonas Hipofisarias/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-3/metabolismo , Células Madre/citología , Tirosina 3-Monooxigenasa/metabolismo
17.
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
18.
Br J Pharmacol ; 112(3): 717-9, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7921594

RESUMEN

Electrophysiological recordings were made from neurones of the medial habenula (Mhb) in brain slices obtained from guinea-pig, rat and gerbil brain. The selective NK3 agonist, senktide, was used to determine the relative number of NK3-sensitive-neurones in the Mhb of each species. The proportion of neurones responding to NK1 (Sar9Met(O2)11SP) and NK2 (beta Ala8NKA(4-10) agonists was also assessed. All (65/65) of the guinea-pig Mhb neurones tested were excited by the NK3 agonist, but NK1 and NK2 agonists were without effect. NK3 responses in the guinea-pig were not altered by the presence of a selective NK1 antagonist, CP-99,994. NK1, NK2 and NK3 agonists were without effect on Mhb neurones from gerbil brain slices. In agreement with findings from previous studies, a population of rat Mhb neurones responded to NK1 or NK3 agonists alone or were excited by both. These data show that there is a difference in both the number of NK-sensitive neurones and the type of NK response found in the medial habenula of the three species. The high sensitivity to an NK3 agonist, combined with the apparent lack of NK1 and NK2 responses in the guinea-pig Mhb makes this preparation ideal for studies of central NK3-mediated events.


Asunto(s)
Neuronas/metabolismo , Receptores de Neuroquinina-3/metabolismo , Tálamo/metabolismo , Animales , Electrofisiología , Gerbillinae , Cobayas , Técnicas In Vitro , Ratas , Ratas Wistar , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/antagonistas & inhibidores , Especificidad de la Especie , Tálamo/citología
19.
Eur J Pharmacol ; 256(1): 73-8, 1994 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-7517891

RESUMEN

[3H]Senktide, a highly selective tachykinin NK3 receptor agonist, was used to study tachykinin NK3 receptors of rat and guinea pig brain. Guinea pig brain membranes had a Kd of 3.9 +/- 0.5 nM and a Bmax of 42 fmol/mg. Dose-displacement experiments with neurokinins and selective tachykinin receptor agonists revealed the following order of potency: [MePhe7]neurokinin B > neurokinin B > substance P > neurokinin A. This order is typical for a tachykinin NK3 receptor. To further characterize the specificity of this receptor, the effects of unrelated compounds such as: bradykinin, angiotensin II, bombesin and their structural analogs were also evaluated on the binding of [3H]senktide. Unexpectedly, the angiotensin AT1 receptor antagonists, DuP 753 (2-n-butyl-4-chloro-5-hydroxymethyl-1-[2'-(1H-tetrazol-5-yl)bip hen yl-4-yl)methyl]imidazole potassium salt), L-158,809 (5,7-dimethyl-2-ethyl-3-[(2'-(1H-tetrazol-5-yl) [1,1'-biphenyl]-4-yl) methyl]-3H-imidazo[4,5-beta]pyridine H2O) and EXP 3174 (2-n-butyl-4-chloro-1-[2'-(1H-tetrazol-5-yl)biphenyl-4-yl)methyl]i midazole- 5-carboxylic acid), inhibited the binding of [3H]senktide to its receptor in the guinea pig brain membranes with IC50 values of 18 microM, 25 microM and 50 microM, respectively. Similar effects were also observed with rat brain membranes. Angiotensin II, saralasin ([Sar1,Val5,Ala8]angiotensin II, a peptide angiotensin AT1 receptor antagonist) and PD 123,319 (1-[4-(dimethylamino)3-methylphenyl]methyl-5-(diphenylacetyl)-4,5, 6,7- tetrahydro-1H-imidazo[4,5-c]pyridine-6-carboxylic acid, a known non-peptide angiotensin AT2 receptor antagonist) did not inhibit the binding of [3H]senktide to either type of membrane.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Antagonistas de Receptores de Angiotensina , Química Encefálica/efectos de los fármacos , Receptores de Neuroquinina-3/metabolismo , Angiotensina II/metabolismo , Animales , Unión Competitiva/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Cobayas , Técnicas In Vitro , Cinética , Ligandos , Membranas/efectos de los fármacos , Membranas/metabolismo , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/efectos de los fármacos , Receptores de Neuroquinina-1/metabolismo , Sustancia P/análogos & derivados , Sustancia P/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA