Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 319: 22-38, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28161095

RESUMEN

Tributyltin chloride (TBT) is a xenobiotic used as a biocide in antifouling paints that has been demonstrated to induce endocrine-disrupting effects, such as obesity and reproductive abnormalities. An integrative metabolic control in the hypothalamus-pituitary-gonadal (HPG) axis was exerted by leptin. However, studies that have investigated the obesogenic TBT effects on the HPG axis are especially rare. We investigated whether metabolic disorders as a result of TBT are correlated with abnormal hypothalamus-pituitary-gonadal (HPG) axis function, as well as kisspeptin (Kiss) action. Female Wistar rats were administered vehicle and TBT (100ng/kg/day) for 15days via gavage. We analyzed their effects on the tin serum and ovary accumulation (as biomarker of TBT exposure), estrous cyclicity, surge LH levels, GnRH expression, Kiss action, fertility, testosterone levels, ovarian apoptosis, uterine inflammation, fibrosis, estrogen negative feedback, body weight gain, insulin, leptin, adiponectin levels, as well as the glucose tolerance (GTT) and insulin sensitivity tests (IST). TBT led to increased serum and ovary tin levels, irregular estrous cyclicity, and decreased surge LH levels, GnRH expression and Kiss responsiveness. A strong negative correlation between the serum and ovary tin levels with lower Kiss responsiveness and GnRH mRNA expression was observed in TBT rats. An increase in the testosterone levels, ovarian and uterine fibrosis, ovarian apoptosis, and uterine inflammation and a decrease in fertility and estrogen negative feedback were demonstrated in the TBT rats. We also identified an increase in the body weight gain and abnormal GTT and IST tests, which were associated with hyperinsulinemia, hyperleptinemia and hypoadiponectinemia, in the TBT rats. TBT disrupted proper functioning of the HPG axis as a result of abnormal Kiss action. The metabolic dysfunctions co-occur with the HPG axis abnormalities. Hyperleptinemia as a result of obesity induced by TBT may be associated with abnormal HPG function. A strong negative correlation between the hyperleptinemia and lower Kiss responsiveness was observed in the TBT rats. These findings provide evidence that TBT leads to toxic effects direct on the HPG axis and/or indirectly by abnormal metabolic regulation of the HPG axis.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas/metabolismo , Leptina/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Compuestos de Trialquiltina/toxicidad , Animales , Disruptores Endocrinos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Hormonas Hipotalámicas/antagonistas & inhibidores , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Kisspeptinas/antagonistas & inhibidores , Leptina/antagonistas & inhibidores , Obesidad/inducido químicamente , Obesidad/metabolismo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Ratas Wistar , Reproducción/efectos de los fármacos , Reproducción/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
2.
Reprod Domest Anim ; 52 Suppl 2: 354-358, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27859771

RESUMEN

The intent of this contribution is to provide an update of the progress we have made towards developing a method/treatment to permanently sterilize cats. Our approach employs two complementary methodologies: RNA interference (RNAi) to silence genes involved in the central control of reproduction and a virus-based gene therapy system intended to deliver RNAi selectively to the hypothalamus (where these genes are expressed) via the systemic administration of modified viruses. We selected the hypothalamus because it contains neurons expressing Kiss1 and Tac3, two genes essential for reproduction and fertility. We chose the non-pathogenic adeno-associated virus (AAV) as a vector whose tropism could be modified to target the hypothalamus. The issues that must be overcome to utilize this vector as a delivery vehicle to induce sterility include modification of the wild-type AAV to target the hypothalamic region of the brain with a simultaneous reduction in targeting of peripheral tissues and non-hypothalamic brain regions, identification of RNAi targets that will effectively reduce the expression of Kiss1 and Tac3 without off-target effects, and determination if neutralizing antibodies to the AAV serotype of choice are present in cats. Successful resolution of these issues will pave the way for the development of a powerful tool to induce the permanent sterility in cats.


Asunto(s)
Gatos , Anticoncepción/veterinaria , Dependovirus , Silenciador del Gen , Vectores Genéticos , Hipotálamo , Animales , Anticoncepción/métodos , Expresión Génica/efectos de los fármacos , Ingeniería Genética/métodos , Ingeniería Genética/veterinaria , Infertilidad/etiología , Infertilidad/veterinaria , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/genética , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/genética , Interferencia de ARN
3.
Biol Reprod ; 93(3): 76, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26246220

RESUMEN

The importance of kisspeptin in regulating vertebrate reproduction has been well established, but the exact mechanism continues to unfold. Unlike mammals, many lower vertebrates possess a dual kisspeptin system, Kiss1 and Kiss2. To decipher the roles of the kisspeptins in fish, we identified two potential kisspeptin antagonists, pep 234 and pep 359, by screening analogs for their ability to inactivate striped bass Kiss1 and Kiss2 receptors expressed in COS7 cells. Pep 234 (a mammalian KISS1 antagonist) antagonizes Kiss1r signaling activated by Kiss1 and Kiss2, and pep 359 (a novel analog) antagonizes Kiss2 activation of both receptors. In vitro studies using brain slices demonstrated that only Kiss2 can upregulate the expression of the hypophysiotropic gnrh1, which was subsequently diminished by pep 234 and pep 359. In primary pituitary cell cultures, the two antagonists revealed a complex network of putative endogenous and exogenous regulation by kisspeptin. While both kisspeptins stimulate Fsh expression and secretion, Kiss2 predominately induces Lh secretion. Pep 234 and 359 treatment of spawning males hindered sperm production. This effect was accompanied with decreased brain gnrh1 and gnrh2 mRNA levels and peptide content in the pituitary, and increased levels of pituitary Lh, probably due to attenuation of Lh release. Strikingly, the mRNA levels of arginine-vasotocin, the neurons of which in the preoptic area coexpress kiss2r, were dramatically reduced by the antagonists. Our results demonstrate differential actions of Kiss1 and Kiss2 systems along the hypothalamic-pituitary axis and interactions with other neuropeptides, and further reinforce the importance of kisspeptin in the execution of spawning.


Asunto(s)
Lubina/genética , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/metabolismo , Reproducción/genética , Animales , Química Encefálica/genética , Células COS , Chlorocebus aethiops , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Liberadora de Gonadotropina/genética , Humanos , Kisspeptinas/genética , Hormona Luteinizante/metabolismo , Masculino , Neuronas/metabolismo , Hipófisis/metabolismo , Cultivo Primario de Células , ARN Mensajero/biosíntesis , Vasotocina/metabolismo
4.
Reproduction ; 149(5): 465-73, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25713426

RESUMEN

The aim of this study was to determine the modulatory effects of peptide 234 (p234) (an antagonist of GPR54 receptors) on kisspeptin and RF9 (an RFamide-related peptide antagonist)-induced changes in reproductive functions and energy balance in female rats. Female Sprague-Dawley rats were weaned on postnatal day (pnd) 21. The animals were intracerebroventricularly cannulated under general anesthesia on pnd 23. Groups of female rats were injected with kisspeptin, RF9, p234, kisspeptin plus p234, or RF9 plus p234, daily. The experiments were ended on the day of first diestrus following pnd 60. Kisspeptin or RF9 alone advanced vaginal opening (VO), which was delayed by administration of kisspeptin antagonist alone. In the rats given kisspeptin plus p234 or RF9 plus p234, VO was not different from control rats. Kisspeptin and RF9 elicited significant elevations in circulating LH levels. Coadministrations of kisspeptin or RF9 with p234 decreased LH levels significantly. The use of p234 alone did not cause any significant change in LH secretion. Kisspeptin decreased both food intake and body weight while RF9 decreased only food intake without affecting body weight. The effects of kisspeptin on energy balance were also reversed by central administration of p234. In conclusion, kisspeptin antagonist, p234, modulates the effects of kisspeptin on reproductive functions and energy balance, whereas RF9 seems to exert only its effects on reproductive functions by means of GPR54 signaling in female rats.


Asunto(s)
Kisspeptinas/antagonistas & inhibidores , Neuropéptidos/farmacología , Péptidos/farmacología , Reproducción/fisiología , Animales , Peso Corporal , Metabolismo Energético , Femenino , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Maduración Sexual/efectos de los fármacos
5.
Neuroendocrinology ; 99(3-4): 190-203, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25011649

RESUMEN

BACKGROUND/AIMS: Kisspeptin is the major excitatory regulator of gonadotropin-releasing hormone (GnRH) neurons and is responsible for basal GnRH/LH release and the GnRH/LH surge. Although it is widely assumed, based on mutations in kisspeptin and Kiss1R, that kisspeptin acts to sustain basal GnRH neuronal activity, there have been no studies to investigate whether endogenous basal kisspeptin tone plays a direct role in basal spontaneous GnRH neuronal excitability. It is also of interest to examine possible interactions between endogenous kisspeptin tone and other neuropeptides that have direct effects on GnRH neurons, such as neuropeptide Y (NPY) or cocaine- and amphetamine-regulated transcript (CART), since the activity of all these neuropeptides changes during states of negative energy balance. METHODS: Loose cell-attached and whole-cell current patch-clamp recordings were made from GnRH-GFP neurons in hypothalamic slices from female and male rats. RESULTS: Kisspeptin activated GnRH neurons in a concentration-dependent manner with an EC50 of 3.32 ± 0.02 nM. Surprisingly, a kisspeptin antagonist, Peptide 347, suppressed spontaneous activity in GnRH neurons, demonstrating the essential nature of the endogenous kisspeptin tone. Furthermore, inhibition of endogenous kisspeptin tone blocked the direct activation of GnRH cells that occurs in response to antagonism of NPY Y5 receptor or by CART. CONCLUSIONS: Our electrophysiology studies suggest that basal endogenous kisspeptin tone is not only essential for spontaneous GnRH neuronal firing, but it is also required for the net excitatory effects of other neuropeptides, such as CART or NPY antagonism, on GnRH neurons. Therefore, endogenous kisspeptin tone could serve as the linchpin in GnRH activation or inhibition.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Kisspeptinas/metabolismo , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Estradiol/farmacología , Femenino , Hormona Liberadora de Gonadotropina/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/farmacología , Masculino , Neuronas/fisiología , Ovariectomía , Técnicas de Placa-Clamp , Área Preóptica/citología , Ratas , Ratas Transgénicas , Ratas Wistar , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
6.
Adv Gerontol ; 27(1): 81-6, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-25051762

RESUMEN

Kisspeptin activates neurocytes and astrocytes of the preoptic hypothalamic nucleus of 1-, 3- and 24-month-old male rats. Kisspeptin antagonist (P-234) depresses the neurocytes, but not the astrocytes of the preoptic nucleus. Melatonin at a dose of 100 mkg/100 g b.w. inhibits the neurons of old male rats. During combined administration of melatonin and kisspeptin, as well as melatonin and P-234, the state of the kisspeptinergic system is crusial for the activity of the neurons in the preoptic nucleus of 1- and 3-month-old animals. However, in old rats melatonin significantly changes the neuron response of the preoptic nucleus to kisspeptin and its antagonist administration, while it's observed the neuron stimulation. Generally, the state of the kisspeptinergic system has a determining influence on the preoptic hypothalamic nucleus of the immature and young mature male rats. In old rats the cell functional state of the preoptic nucleus depends on the interaction of the kisspeptinergic system and melatonin level.


Asunto(s)
Astrocitos , Hipotálamo , Kisspeptinas , Melatonina , Neuronas , Factores de Edad , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biotransformación , Senescencia Celular/fisiología , Vías de Administración de Medicamentos , Hipotálamo/metabolismo , Hipotálamo/patología , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Masculino , Melatonina/metabolismo , Melatonina/farmacología , Neuronas/metabolismo , Neuronas/patología , Área Preóptica/metabolismo , Área Preóptica/patología , Ratas , Ratas Wistar , Resultado del Tratamiento
7.
Adv Exp Med Biol ; 784: 159-86, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23550006

RESUMEN

Kisspeptin is now known to be an important regulator of the hypothalamic--pituitary-gonadal axis and is the target of a range of regulators, such as steroid hormone feedback, nutritional and metabolic regulation. Kisspeptin binds to its cognate receptor, KISS1R (also called GPR54), on GnRH neurons and stimulates their activity, which in turn provides an obligatory signal for GnRH secretion-thus gating down-stream events supporting reproduction. The development of peripherally active kisspeptin antagonists could offer a unique therapeutic agent for treating hormone-dependent disorders of reproduction, including precocious puberty, endometriosis, and metastatic prostate cancer. The following chapter discusses the advances made in the search for both peptide and small molecule kisspeptin antagonists and their use in delineating the role of kisspeptin within the reproductive system. To date, four peptide antagonists and one small molecule antagonist have been designed.


Asunto(s)
Endometriosis/tratamiento farmacológico , Infertilidad/tratamiento farmacológico , Kisspeptinas/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Pubertad Precoz/tratamiento farmacológico , Animales , Endometriosis/patología , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Infertilidad/patología , Masculino , Neuronas/metabolismo , Neuronas/patología , Neoplasias de la Próstata/patología , Pubertad Precoz/patología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Reproducción
8.
J Clin Endocrinol Metab ; 107(1): e328-e347, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34387319

RESUMEN

CONTEXT: Recent evidence suggests that vasomotor symptoms (VMS) or hot flashes in the postmenopausal reproductive state and polycystic ovary syndrome (PCOS) in the premenopausal reproductive state emanate from the hyperactivity of Kiss1 neurons in the hypothalamic infundibular/arcuate nucleus (KNDy neurons). OBJECTIVE: We demonstrate in 2 murine models simulating menopause and PCOS that a peripherally restricted kappa receptor agonist (PRKA) inhibits hyperactive KNDy neurons (accessible from outside the blood-brain barrier) and impedes their downstream effects. DESIGN: Case/control. SETTING: Academic medical center. PARTICIPANTS: Mice. INTERVENTIONS: Administration of peripherally restricted kappa receptor agonists and frequent blood sampling to determine hormone release and body temperature. MAIN OUTCOME MEASURES: LH pulse parameters and body temperature. RESULTS: First, chronic administration of a PRKA to bilaterally ovariectomized mice with experimentally induced hyperactivity of KNDy neurons reduces the animals' elevated body temperature, mean plasma LH level, and mean peak LH per pulse. Second, chronic administration of a PRKA to a murine model of PCOS, having elevated plasma testosterone levels and irregular ovarian cycles, suppresses circulating levels of LH and testosterone and restores normal ovarian cyclicity. CONCLUSION: The inhibition of kisspeptin neuronal activity by activation of kappa receptors shows promise as a novel therapeutic approach to treat both VMS and PCOS in humans.


Asunto(s)
Sofocos/tratamiento farmacológico , Kisspeptinas/antagonistas & inhibidores , Menopausia/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Receptores Opioides kappa/agonistas , Animales , Buprenorfina/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Sofocos/sangre , Sofocos/etiología , Humanos , Kisspeptinas/metabolismo , Meloxicam/administración & dosificación , Menopausia/sangre , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Receptores Opioides kappa/metabolismo , Sistema Vasomotor/efectos de los fármacos
9.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31619585

RESUMEN

During pregnancy the maternal pancreatic islets of Langerhans undergo adaptive changes to compensate for gestational insulin resistance. Kisspeptin has been shown to stimulate insulin release, through its receptor, GPR54. The placenta releases high levels of kisspeptin into the maternal circulation, suggesting a role in modulating the islet adaptation to pregnancy. In the present study we show that pharmacological blockade of endogenous kisspeptin in pregnant mice resulted in impaired glucose homeostasis. This glucose intolerance was due to a reduced insulin response to glucose as opposed to any effect on insulin sensitivity. A ß cell-specific GPR54-knockdown mouse line was found to exhibit glucose intolerance during pregnancy, with no phenotype observed outside of pregnancy. Furthermore, in pregnant women circulating kisspeptin levels significantly correlated with insulin responses to oral glucose challenge and were significantly lower in women with gestational diabetes (GDM) compared with those without GDM. Thus, kisspeptin represents a placental signal that plays a physiological role in the islet adaptation to pregnancy, maintaining maternal glucose homeostasis by acting through the ß cell GPR54 receptor. Our data suggest reduced placental kisspeptin production, with consequent impaired kisspeptin-dependent ß cell compensation, may be a factor in the development of GDM in humans.


Asunto(s)
Diabetes Gestacional/fisiopatología , Intolerancia a la Glucosa/fisiopatología , Células Secretoras de Insulina/fisiología , Kisspeptinas/metabolismo , Placenta/metabolismo , Adaptación Fisiológica , Adulto , Animales , Diabetes Gestacional/sangre , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/metabolismo , Femenino , Glucosa/análisis , Glucosa/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/diagnóstico , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/administración & dosificación , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/sangre , Ratones , Ratones Noqueados , Ratones Transgénicos , Placenta/efectos de los fármacos , Circulación Placentaria/fisiología , Embarazo , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo
10.
Theriogenology ; 121: 134-140, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30149259

RESUMEN

Kisspeptin is the protein product of Kiss1 gene. In contrast to its fundamental role as GnRH Secretagogue and puberty initiation, its functions in extrahypothalmic tissues such as the testes remain unknown. The objective of this study was to investigate the potential local role of kisspeptin in the Leydig cell culture in goats. Five male Shiba goats were castrated to isolate Leydig cells from the testes. The isolated Leydig cells were then purified and cultured with different concentration of kisspeptin antagonist (0, 5, and 10 nmol; 0KPA, 5KPA, and 10KPA; respectively). After 12 h, 20 IU of hCG was added to some wells (0KPA + hCG, 5KPA + hCG, and 10KPA + hCG; respectively); other wells did not receive hCG (0KPA + Con, 5KPA + Con, and 10KPA + Con; respectively). Media of Leydig cell culture was harvested at 2, 6, 12, and 24 h after addition of hCG for testosterone (T) and estradiol (E2) assays. The contents of some non KPA treated wells (0KPA + Con and 0KPA + hCG wells) were collected and stored at -80 OC for further identification the expression of mRNA encoding Kiss1 and its receptor (GPR54) using real time PCR. Results revealed high expressions of Kiss1/GPR54 mRNAs in the hCG treated (0KPA + hCG) wells compared to non-hCG treated ones (0KPA + Con). KPA significantly reduced the basal T production at 6 h incubation as well as hCG-induced T production in 2, 6, and 24 h incubation. Basal E2 concentrations were significantly lower in the KPA treated wells at 6, and 24 h likewise, KPA significantly attenuated induced E2 production at 12 h compared with its values in non KPA treated wells. In conclusion, kisspeptin antagonist significantly attenuated both basal and hCG-activated T and E2 production by purified Leydig cells in goats. Therefore, involvement of kisspeptin in the steroidogenic capability of Leydig cells is possible in goats.


Asunto(s)
Cabras , Hormonas Esteroides Gonadales/metabolismo , Kisspeptinas/antagonistas & inhibidores , Células Intersticiales del Testículo/efectos de los fármacos , Animales , Células Intersticiales del Testículo/metabolismo , Masculino
11.
J Endocrinol ; 237(2): 165-173, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29549187

RESUMEN

Kisspeptin signalling is indispensable for fertility, stimulating gonadotropin-releasing hormone (GnRH) secretion and mediating gonadal steroid feedback on GnRH neurons. Moreover, kisspeptin neurons have been implicated in other non-reproductive neuroendocrine roles. Kisspeptin appears to also regulate growth hormone secretion but much of the data appear contradictory. We sought to clarify a potential role of kisspeptin in growth hormone (GH) regulation by examining the effect of kisspeptin antagonists on GH secretion in ewes under various physiological conditions. Our data show clear and robust increases in GH secretion following lateral ventricle or third ventricle infusion of kisspeptin antagonists p-234 and p-271 in either ovariectomized or anestrous ewes. Central infusion of kisspeptin-10 had no effect on GH secretion. To determine the level at which kisspeptin may influence GH secretion, we examined expression of the cognate kisspeptin receptor, GPR54, in pituitary cells and showed by immunocytochemistry that the majority of somatotropes express GPR54 while expression was largely negative in other pituitary cells. Overall, we have demonstrated that blocking kisspeptin signalling by antagonists stimulates GH secretion in ewes and that this is likely mediated by inhibiting endogenous kisspeptin activation of GPR54 expressed on somatotropes. The findings suggest that endogenous kisspeptin inhibits GH secretion through GPR54 expressed on somatotropes.


Asunto(s)
Hormona del Crecimiento/metabolismo , Antagonistas de Hormonas/farmacología , Kisspeptinas/antagonistas & inhibidores , Animales , Femenino , Hormona del Crecimiento/sangre , Hidrocortisona/metabolismo , Infusiones Intraventriculares , Kisspeptinas/administración & dosificación , Kisspeptinas/farmacología , Hormona Luteinizante/metabolismo , Ovariectomía , Ovario/fisiología , Prolactina/metabolismo , Vías Secretoras/efectos de los fármacos , Ovinos
12.
Peptides ; 105: 21-27, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29709623

RESUMEN

Kisspeptin, a hypothalamic neuropeptide, is a member of the RF-amide family, which have been known to modify pain sensitivity in rodents. The aim of the present study was to investigate the effect of kisspeptin-13 (KP-13), an endogenous derivative of kisspeptin, on nociception in adult male and female CFLP mice and the possible interaction of KP-13 with morphine on nociception. Mice were injected with different doses of KP-13, 30, 60 and 120 min after of which the nociceptive sensitivity were assessed via the tail-flick test. To investigate the receptor involved in the mediation a kisspeptin receptor antagonist (KP-234) pretreatment was applied before KP-13 administration. Furthermore, we investigated the effect of KP-13 on the acute antinociceptive effect of morphine, on acute morphine tolerance and on naloxone-precipitated withdrawal. Last, the Von Frey test was used in order to assess KP-13's effect on mechanical nociception. Our results showed that KP-13 decreased the nociceptive threshold of both males and females independent of sex, which was prevented by KP-234. Furthermore, KP-13 treatment depressed the acute antinociceptive effect of morphine and attenuated the development of morphine tolerance. KP-13 also induced a mechanical hypersensitivity. These data underlie kisspeptin's hyperalgesic action and argues for the role of kisspeptin receptor 1 in the mediation of its action. Furthermore, our results suggest that central KP-13 administration can modify the acute effects of morphine.


Asunto(s)
Tolerancia a Medicamentos/genética , Kisspeptinas/genética , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Analgésicos Opioides/administración & dosificación , Animales , Kisspeptinas/antagonistas & inhibidores , Ratones , Morfina/administración & dosificación , Nocicepción/fisiología , Dolor/genética , Dolor/fisiopatología , Umbral del Dolor/efectos de los fármacos , Receptores de Kisspeptina-1/antagonistas & inhibidores
13.
J Physiol Pharmacol ; 69(3)2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30342431

RESUMEN

In this study, we tested the hypothesis that modulation of endogenous gonadotropin-releasing hormone (Gnrh) neuronal network activity alters the mRNA expression of nuclear receptor subfamily 5 group A member 1 (Nr5a1), through one of the component of Wnt pathway signaling - catenin beta 1 (Ctnnb1) (its co-activator), and its co-repressor nuclear receptor subfamily 0, group B member 1 (Nr0b1) in the female rat pituitary gland in vivo. Adult ovariectomized rats were given a serial infusion of Gnrh, kisspeptin-10, Gnrh + Gnrh antagonist (Antide), or kisspeptin-10 + kisspeptin antagonist (kisspeptin-234) into the third ventricle of the brain. The anterior pituitary and blood was used to mRNA and protein expression analysis. We demonstrated that Gnrh up-regulates Nr5a1 mRNA expression in the anterior pituitary and induces NR5A1 depletion in gonadotropes. Gnrh administration increased both Ctnnb1 mRNA expression and protein synthesis, and induced activation of cellular Ctnnb1 via translocation from the gonadotropes cytoplasm to nucleus. After kisspeptin-10 treatment, up-regulation of Nr0b1 mRNA and protein expression in the anterior pituitary was observed. These data indicate that Gnrh-neuron-mediated network activity alters Nr5a1 gene transcription and translation in gonadotrope cells and this effect may result from the changes induced in the Ctnnb1 and Nr0b1 gene/protein expression balance.


Asunto(s)
Receptor Nuclear Huérfano DAX-1/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Kisspeptinas/farmacología , Adenohipófisis/efectos de los fármacos , Factor Esteroidogénico 1/metabolismo , beta Catenina/metabolismo , Animales , Receptor Nuclear Huérfano DAX-1/genética , Femenino , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Kisspeptinas/antagonistas & inhibidores , Adenohipófisis/metabolismo , Ratas Wistar , Factor Esteroidogénico 1/genética , beta Catenina/genética
14.
Theriogenology ; 115: 1-8, 2018 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-29698886

RESUMEN

Kisspeptin and its receptor KISS1R are involved in the neuroendocrine regulation of mammalian reproduction and their role on follicular development and function can be hypothesized. The present work was designed to confirm the immunopresence of kisspeptin and its receptor in the ovary of swine and to study the effects of kisspeptin 10 and its antagonist, kisspeptin 234, on main functional parameters of granulosa cells (i.e. cell proliferation, steroid production, and redox status) as well as their modulatory action on angiogenesis. The immunopresence of kisspeptin and KISS1R were detected in granulosa cells. Kisspeptin 10 stimulated progesterone in vitro production, thus indirectly suggesting that it can have a role in the luteinization process of granulosa cells. Kisspeptin 10 displayed potentiating effects on non-enzymatic scavenging activity, thus supporting its involvement in the control of the antioxidant defense system of ovarian follicles. In addition, results from the angiogenesis bioassay suggest that kisspeptin may have a role in the physiological development of new ovarian vessels. Additional studies are needed to confirm the functional significance of the kisspeptin/KISS1R system within the swine ovary.


Asunto(s)
Kisspeptinas/fisiología , Folículo Ovárico/química , Folículo Ovárico/fisiología , Receptores de Kisspeptina-1/fisiología , Porcinos , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Células de la Granulosa/química , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/fisiología , Kisspeptinas/análisis , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Ovario/irrigación sanguínea , Ovario/fisiología , Oxidación-Reducción , Progesterona/biosíntesis , Receptores de Kisspeptina-1/análisis
15.
Artículo en Inglés | MEDLINE | ID: mdl-29729480

RESUMEN

Semicarbazide (SMC), a new marine pollutant, has anti-estrogenic effects on female Japanese flounder (Paralichthys olivaceus). However, whether SMC also affects the reproductive endocrine system of male marine organisms is currently unclear. In this study, Japanese flounder embryos were exposed to 1, 10, and 100 µg/L SMC for 130 days. Plasma testosterone (T) and 17ß-estradiol (E2) concentrations were significantly decreased in male flounders after SMC exposure. The expression of genes involved in T and E2 synthesis, including steroidogenic acute regulatory protein, cytochrome P450 11A1, 17α-hydroxylase, 17ß-hydroxysteroid dehydrogenase and cytochrome P450 19A, was down-regulated in the gonads, which may explain the decrease in plasma sex hormones levels. Moreover, SMC-mediated changes in the transcription of these steroidogenic genes were associated with reduced levels of follicle-stimulating hormone beta subunit (fshß), luteinizing hormone beta subunit (lhß), follicle-stimulating hormone receptor (fshr) and luteinizing hormone receptor (lhr) mRNA. In addition, down-regulated transcription of fshß and lhß in the SMC exposure groups was affected by reduced mRNA levels of seabream gonadotropin-releasing hormone (sbgnrh), g-protein-coupled receptor 54 (gpr54) in the kisspeptin/gpr54 system, as well as the gamma-aminobutyric acid (GABA) synthesis enzyme glutamic acid decarboxylase (gad). Overall, our results showed that environmentally relevant concentrations of SMC exerted anti-androgenic effects in male flounders via impacting HPG axis, kiss/gpr54 system and GABA synthesis, providing theoretical support for investigating reproductive toxicity of environmental pollutants that interfere with the neuroendocrine system.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Peces Planos/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Semicarbacidas/toxicidad , Testículo/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Antagonistas de Andrógenos/toxicidad , Animales , Acuicultura , China , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Represión Enzimática/efectos de los fármacos , Estradiol/biosíntesis , Estradiol/sangre , Estradiol/química , Antagonistas de Estrógenos/toxicidad , Proteínas de Peces/antagonistas & inhibidores , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Peces Planos/sangre , Peces Planos/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Distribución Aleatoria , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testosterona/antagonistas & inhibidores , Testosterona/biosíntesis , Testosterona/sangre , Pruebas de Toxicidad Crónica
16.
Cell Signal ; 42: 1-10, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28988968

RESUMEN

The invasive and metastatic phenotypes of breast cancer correlate with high recurrence rates and poor survival outcomes. Transforming growth factor-ß (TGFß) promotes tumor progression and metastasis in aggressive breast cancer. Here, we identified the kisspeptin KiSS1 as a downstream target of canonical TGFß/Smad2 pathway in triple negative breast cancer cells. We also found KiSS1 expression to be required for TGFß-induced cancer cell invasion. Indeed, knockdown expression of KiSS1 blocked TGFß-mediated cancer cell invasion as well as metalloproteinase (MMP9) expression and activity. Interestingly, Kisspeptin-10 (KP-10), the smallest active form of kisspeptin also stimulates cancer cell invasive behavior through activation of MAPK/Erk pathway. We described a positive feedback loop between KiSS1 and p21 downstream of TGFß, further contributing to TGFß-induced cancer cell invasion. Lastly, we explored both the clinical utility of KiSS1 as a lymph node involvement predictive tool and its potential as a therapeutic target. We found KiSS1 high expression to correlate with lymph node positive status. Furthermore, blocking KiSS1 using a specific small peptide antagonist (p234) impaired TGFß-mediated cell invasion and MMP9 induction. Together, our results define an essential role of KiSS1 in regulating TGFß pro-invasive effects and define KiSS1 as a therapeutic new target for triple negative breast cancer.


Asunto(s)
Adenocarcinoma/genética , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Kisspeptinas/genética , Factor de Crecimiento Transformador beta/genética , Neoplasias de la Mama Triple Negativas/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Metástasis Linfática , Células MCF-7 , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama Triple Negativas/diagnóstico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
17.
J Endocrinol ; 233(2): 159-174, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28330973

RESUMEN

Kisspeptin and neurokinin B (NKB) are neuropeptides co-expressed in the mammalian hypothalamus and coordinately control GnRH signaling. We have found that Nkb and kisspeptin neurons are distinct in the teleost, striped bass (STB) and capitalized on this phenomenon to study the mode of action of Nkb and its related neuropeptide-F (Nkf), both of which are encoded by the tac3 gene. In vitro brain slices and in vivo administration studies revealed that Nkb/f consistently downregulated kiss2, whereas antagonist (AntD) administration restored this effect. Overall, a minor effect was noted on gnrh1 expression, whereas Gnrh1 content in the pituitaries was reduced after Nkb/f treatment and increased with AntD. Concomitantly, immunostaining demonstrated that hypothalamic Nkb neurons border and densely innervate the largest kiss2 neuronal population in the hypothalamus, which also coexpresses Nkb receptor. No expression of Nkb receptor or Nkb neuronal projections was detected near/in Gnrh1 soma in the preoptic area. At the level of the pituitary, however, the picture was more complex: both Nkb/f and AntD upregulated lhb and fshb expression and Lh secretion in vivo Together with the stimulatory effect of Nkb/f on Lh/Fsh secretion from pituitary cells, in vitro, this may indicate an additional independent action of Nkb/f within the pituitary, in which the hypothalamic pathway is more dominant. The current study demonstrates that Nkb/f utilizes multiple pathways to regulate reproduction in the STB and that in the brain, Nkb mainly acts as a negative modulator of kiss2 to regulate the release of Gnrh1.


Asunto(s)
Lubina/metabolismo , Regulación de la Expresión Génica/fisiología , Kisspeptinas/metabolismo , Neuroquinina B/fisiología , Reproducción/fisiología , Animales , Clonación Molecular , ADN Complementario/metabolismo , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/genética , Masculino , Neuroquinina B/genética , Hipófisis/metabolismo
18.
Endocrinology ; 158(10): 3269-3280, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977601

RESUMEN

Loss-of-function or inactivating mutations in the genes coding for kisspeptin and its receptor (KISS1R) or neurokinin B (NKB) and the NKB receptor (NK3R) in humans result in a delay in or the absence of puberty. However, precise mechanisms of kisspeptin and NKB signaling in the regulation of the pubertal increase in gonadotropin-releasing hormone (GnRH) release in primates are unknown. In this study, we conducted a series of experiments infusing agonists and antagonists of kisspeptin and NKB into the stalk-median eminence, where GnRH, kisspeptin, and NKB neuroterminal fibers are concentrated, and measuring GnRH release in prepubertal and pubertal female rhesus monkeys. Results indicate that (1) similar to those previously reported for GnRH stimulation by the KISS1R agonist (i.e., human kisspeptin-10), the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in both prepubertal and pubertal monkeys; (2) the senktide-induced GnRH release was blocked in the presence of the KISS1R antagonist peptide 234 in pubertal but not prepubertal monkeys; and (3) the kisspeptin-induced GnRH release was blocked in the presence of the NK3R antagonist SB222200 in the pubertal but not prepubertal monkeys. These results are interpreted to mean that although, in prepubertal female monkeys, kisspeptin and NKB signaling to GnRH release is independent, in pubertal female monkeys, a reciprocal signaling mechanism between kisspeptin and NKB neurons is established. We speculate that this cooperative mechanism by the kisspeptin and NKB network underlies the pubertal increase in GnRH release in female monkeys.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/fisiología , Macaca mulatta/fisiología , Neuroquinina B/fisiología , Maduración Sexual/fisiología , Transducción de Señal/fisiología , Animales , Femenino , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/farmacología , Eminencia Media/efectos de los fármacos , Neuroquinina B/agonistas , Neuroquinina B/antagonistas & inhibidores , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Quinolinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Kisspeptina-1 , Receptores de Neuroquinina-3/agonistas , Transducción de Señal/efectos de los fármacos , Sustancia P/análogos & derivados , Sustancia P/farmacología
19.
Ann Endocrinol (Paris) ; 78 Suppl 1: S31-S40, 2017 Oct.
Artículo en Francés | MEDLINE | ID: mdl-29157487

RESUMEN

The endocrine and exocrine functions of the gonads are controlled by the gonadotrope axis, whose master regulator is the hypothalamic decapeptide GnRH. The Kisspeptin/Neurokinin B (Kp/NkB) neuronendocrine system is the main physiologic regulator of GnRH neurons. The Kp/NkB system is currently considered the key mediator for the hypothalamic negative feedback exerted by sex steroids and prolactin, as well as by various metabolic signals. Intrinsic alterations or regulatory abnormalities of Kp/NkB system lead to various gonadotrope axis puberty and fertility dysfunctions. Molecular inactivations of Kp/NkB system actors are associated with some forms of congenital hypogonadotropic hypogonadism without anosmia. The Kp/NkB System is also involved in a few forms of precocious puberty. Finally, the Kp/NKB system is also implicated in gonadotrope axis alterations leading to functional hypothalamic amenorrhea or hyperprolactinemia. NkB is particularly and directly involved in vasomotor menopausal hot flushes mechanism. Various Kp/NkB agonist/antagonist compounds have been developed during the last ten years, and are currently being evaluated in humans. These molecules have potential applications not only in rare genetic diseases with Kp/NkB alterations, but also in various gonadotrope axis-related diseases or in vitro fertilization. The administration of NkB antagonists in menopausal women represents a real therapeutic advance because of their impressive effect in controlling vasomotor menopausal hot flushes.


Asunto(s)
Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/uso terapéutico , Gónadas/fisiología , Antagonistas de Hormonas/uso terapéutico , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Animales , Femenino , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Humanos , Hipogonadismo/terapia , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/metabolismo , Masculino , Menopausia/efectos de los fármacos , Neuroquinina B/agonistas , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/metabolismo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Maduración Sexual/efectos de los fármacos , Maduración Sexual/fisiología
20.
Endocr Dev ; 30: 106-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26684214

RESUMEN

Reproductive hormones play a role at all stages of life and affect most tissues of the body. Gonadotropin-releasing hormone (GnRH) synthesized in the hypothalamus stimulates the secretion of gonadotropins which in turn stimulate gonadal sex hormone production and gamete formation. This hypothalamic-pituitary-gonadal (HPG) axis has, therefore, been the target for the development of numerous drugs which regulate it at various points. These include sex steroid agonists and antagonists, inhibitors of sex steroid biosynthesis, and GnRH agonists and antagonists, which have found extensive applications in treating numerous conditions such as precocious puberty, delayed puberty, prostate cancer, benign prostatic hyperplasia, endometriosis, uterine fibroids and also in in vitro fertilization protocols. The novel neuroendocrine peptides, kisspeptin (KP) and neurokinin B (NKB), were recently discovered as upstream regulators of GnRH, and inactivating mutations of KP and NKB ligands or receptors result in a failure to progress through puberty. Agonists and antagonists of KP and NKB are being developed as more subtle modulators of the HPG axis. These new drugs offer additional and alternative therapeutic options in pediatric and adult hormone-dependent diseases.


Asunto(s)
Hormonas Esteroides Gonadales , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gónadas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas , Neuroquinina B , Hormonas Esteroides Gonadales/agonistas , Hormonas Esteroides Gonadales/antagonistas & inhibidores , Humanos , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inhibidores , Neuroquinina B/agonistas , Neuroquinina B/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA