Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Rev Endocrinol ; 16(8): 407-420, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32427949

RESUMEN

Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.


Asunto(s)
Metabolismo Energético/fisiología , Kisspeptinas/fisiología , Reproducción/fisiología , Animales , Dinorfinas/fisiología , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Homeostasis , Humanos , Sistema Hipotálamo-Hipofisario/fisiología , Hipotálamo/citología , Hipotálamo/fisiología , Kisspeptinas/genética , Hormona Luteinizante/fisiología , Neuroquinina B/fisiología , Neuronas/fisiología , Ovario/fisiología , Pubertad/fisiología
2.
Gynecol Endocrinol ; 34(11): 913-919, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29902942

RESUMEN

Women during perimenopausal period experience a range of symptoms, which interfere with physical, sexual, and social life. About 65-75% of symptoms connected with postmenopausal period are vasomotor symptoms (VMS), such as hot flushes and night sweats. Hot flushes are subjective sensation of heat associated with cutaneous vasodilatation and drop in core temperature. It is suspected that VMS are strongly correlated with pulsatile oversecretion of gonadotropin-releasing hormone (GnRH) and subsequently luteinizing hormone (LH). Evidence has accumulated in parallel showing that lack of negative feedback of steroid hormones synthesized in ovary causes overactivation of hypertrophied kisspeptin/neurokinin B/dynorphin (KNDy) neurons, located in infundibular nucleus. Oversecretion of both kisspeptin (KISS1) and neurokinin B (NKB), as well as downregulation of dynorphin, plays dominant role in creation of GnRH pulses. This in turn causes VMS. Administration of senktide, highly potent and selective NK3R agonist, resulted in increase of serum LH concentration, induction of VMS, increase in heart rate, and skin temperature in postmenopausal women. These finding suggest that modulation of KNDy neurons may become new therapeutic approach in the treatment of VMS.


Asunto(s)
Sofocos/etiología , Hipotálamo/fisiología , Neuronas/fisiología , Posmenopausia/fisiología , Sistema Vasomotor/fisiología , Dinorfinas/fisiología , Retroalimentación Fisiológica , Femenino , Sofocos/tratamiento farmacológico , Humanos , Kisspeptinas/fisiología , Neuroquinina B/fisiología
3.
Neurobiol Aging ; 50: 30-38, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27842268

RESUMEN

Pulsatile secretion of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) decreases during aging. Kisspeptin (encoded by Kiss1) neurons in the arcuate nucleus coexpress neurokinin B (Tac3) and dynorphin (Pdyn) and are critical for regulating the GnRH/LH pulse. We therefore examined kisspeptin neurons by histochemistry and pulsatile LH release in rats aged 2-3 (Young), 12-13 (Young-Middle), 19-22 (Late-Middle), and 24-26 (Old) months. Total LH concentrations, sampled for 3 hours, decreased in both sexes with aging. In females, numbers of Tac3 and Pdyn neurons were significantly reduced in all aging rats, and numbers of Kiss1 neurons were significantly reduced in Late-Middle and Old rats. In males, numbers of all 3 neuron-types were significantly decreased in all aging rats. GnRH agonist induced LH release in all animals; however, the increased LH concentration in all aging rats was less than that in Young rats. These results suggest that expression of each gene in kisspeptin neurons may be controlled individually during aging, and that reduction of their expression or change in pituitary responsiveness may cause attenuated pulsatile LH secretion.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Dinorfinas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Neuronas/patología , Animales , Dinorfinas/fisiología , Femenino , Histocitoquímica , Hipotálamo/citología , Hipotálamo/patología , Kisspeptinas/fisiología , Masculino , Menopausia/metabolismo , Menopausia/fisiología , Neuroquinina B/fisiología , Neuronas/fisiología , Flujo Pulsátil , Ratas Wistar
4.
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
5.
Phytother Res ; 27(10): 1564-71, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23339028

RESUMEN

Although Berberine (BER) is popular in treating gastrointestinal (GI) disorders, its mechanisms are not clear yet. In order to investigate the effects and possible mechanism of BER on GI motility in rodents, we first explored GI motility by recording the myoelectrical activity of jejunum and colon in rats, and upper GI transit with a charcoal marker in mice. Then, the plasma levels of gastrin, motilin, somatostatin and glucagon-like-peptide-1 (Glp-1) were measured by ELISA or radioimmunoassay (RIA). Furthermore, endogenous opioid-peptides (ß-endorphin, dynorphin-A, met-enkephalin) were detected by RIA after treatment with BER. Our results showed that BER concentration-dependently inhibited myoelectrical activity and GI transit, which can be antagonized by opioid-receptor antagonists to different extents. The elevated somatostatin and Glp-1, and decreased gastrin and motilin in plasma, which were caused by BER application, also could be antagonized by the opioid-receptor antagonists. Additionally, plasma level of ß-endorphin, but not dynorphin-A and met-enkephalin, was increased by applying BER. Taken together, these studies show that BER plays inhibiting roles on GI motility and up-regulating roles on somatostatin, Glp-1 and down-regulating roles on gastrin, motilin. The pharmacological mechanisms of BER on GI motility and plasma levels of GI hormones were discovered to be closely related to endogenous opioid system.


Asunto(s)
Berberina/farmacología , Hormonas Gastrointestinales/fisiología , Motilidad Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Péptidos Opioides/fisiología , Animales , Colon/efectos de los fármacos , Colon/fisiología , Dinorfinas/fisiología , Encefalina Metionina/fisiología , Gastrinas/fisiología , Tracto Gastrointestinal/fisiología , Tránsito Gastrointestinal/efectos de los fármacos , Tránsito Gastrointestinal/fisiología , Péptido 1 Similar al Glucagón/fisiología , Yeyuno/efectos de los fármacos , Yeyuno/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Motilina/fisiología , Ratas , Ratas Sprague-Dawley , Somatostatina/fisiología , betaendorfina/fisiología
6.
Addict Biol ; 18(4): 678-88, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22994904

RESUMEN

The brain stress-response system is critically involved in the addiction process, stimulating drug consumption and the relapse to drug taking in abstinent addicts. At the same time, its functioning is affected by chronic drug exposure. Here, we have investigated the role of the endogenous opioid peptide dynorphin as a modulator of effects of long-term ethanol consumption on the brain stress-response system. Using the two-bottle choice paradigm, we demonstrate an enhanced ethanol preference in male dynorphin knockout mice. Exposure to mild foot shock increased ethanol consumption in wild-type control littermates, but not in dynorphin-deficient animals. Blood adrenocorticotropic hormone levels determined 5 minutes after the shock were not affected by the genotype. We also determined the neuronal reactivity after foot shock exposure using c-Fos immunoreactivity in limbic structures. This was strongly influenced by both genotype and chronic ethanol consumption. Long-term alcohol exposure elevated the foot shock-induced c-Fos expression in the basolateral amygdala in wild-type animals, but had the opposite effect in dynorphin-deficient mice. An altered c-Fos reactivity was also found in the periventricular nucleus, the thalamus and the hippocampus of dynorphin knockouts. Together these data suggest that dynorphin plays an important role in the modulation of the brain stress-response systems after chronic ethanol exposure.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Dinorfinas/fisiología , Etanol/farmacología , Sistema Límbico/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Fisiológico/fisiología , Adaptación Fisiológica/efectos de los fármacos , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Análisis de Varianza , Animales , Conducta Adictiva/metabolismo , Dinorfinas/genética , Etanol/administración & dosificación , Femenino , Preferencias Alimentarias , Genotipo , Inmunoquímica , Sistema Límbico/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados/genética , Refuerzo en Psicología , Autoadministración , Caracteres Sexuales , Estrés Fisiológico/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/metabolismo
7.
Neuropeptides ; 46(6): 383-94, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23062312

RESUMEN

Chronic opiate usage, whether prescribed or illicit, has been associated with changes in bone mass and is a recognized risk factor for the development of osteoporosis; however, the mechanism behind this effect is unknown. Here we show that lack of dynorphin, an endogenous opioid, in mice (Dyn-/-), resulted in a significantly elevated cancellous bone volume associated with greater mineral apposition rate and increased resorption indices. A similar anabolic phenotype was evident in bone of mice lacking dynorphin's cognate receptor, the kappa opioid receptor. Lack of opioid receptor expression in primary osteoblastic cultures and no change in bone cell function after dynorphin agonist treatment in vitro indicates an indirect mode of action. Consistent with a hypothalamic action, central dynorphin signaling induces extracellular signal-regulated kinase (ERK) phosphorylation and c-fos activation of neurons in the arcuate nucleus of the hypothalamus (Arc). Importantly, this signaling also leads to an increase in Arc NPY mRNA expression, a change known to decrease bone formation. Further implicating NPY in the skeletal effects of dynorphin, Dyn-/-/NPY-/- double mutant mice showed comparable increases in bone formation to single mutant mice, suggesting that dynorphin acts upstream of NPY signaling to control bone formation. Thus the dynorphin system, acting via NPY, may represent a pathway by which higher processes including stress, reward/addiction and depression influence skeletal metabolism. Moreover, understanding of these unique interactions may enable modulation of the adverse effects of exogenous opioid treatment without directly affecting analgesic responses.


Asunto(s)
Huesos/fisiología , Dinorfinas/fisiología , Homeostasis/fisiología , Animales , Western Blotting , Composición Corporal/genética , Composición Corporal/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/aislamiento & purificación , Dinorfinas/genética , Femenino , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Neuropéptido Y/fisiología , Osteoblastos/fisiología , Embarazo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Células del Estroma/fisiología , Tomografía Computarizada por Rayos X
8.
Brain Res ; 1364: 103-15, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20951683

RESUMEN

Gonadotropin-releasing hormone (GnRH) secretion has two modes of release in mammalian species; the surge mode and the pulse mode. The surge mode, which is required for the induction of the preovulatory gonadotropin discharge in most species, is induced by the positive feedback of estrogen secreted by the mature ovarian follicle. The pulse mode of GnRH secretion stimulates tonic luteinizing hormone (LH) and follicle-stimulating hormone (FSH) secretion which drives folliculogenesis, spermatogenesis and steroidogenesis and is negatively fine-tuned by estrogen or androgen. The GnRH pulse-generating mechanism is sensitive to environmental cues, such as photoperiod, nutrition and stress surge-generating mechanism is relatively emancipated from these environmental cues. The present article first provides a brief historical background to the work that led to the concept of the GnRH pulse generator: a hypothalamic network that is central to our understanding of the regulation of reproduction. We then discuss possible neurobiological mechanisms underlying GnRH pulse generation, and conclude by proposing that kisspeptin neurons in the arcuate nucleus are key players in this regard.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/fisiología , Neurobiología , Animales , Dinorfinas/fisiología , Ambiente , Hormona Folículo Estimulante/fisiología , Humanos , Kisspeptinas , Hormona Luteinizante/metabolismo , Neuroquinina B/fisiología , Neuronas/fisiología , Esteroides/fisiología , Proteínas Supresoras de Tumor/fisiología
9.
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
10.
Alcohol Clin Exp Res ; 34(5): 761-70, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20184566

RESUMEN

BACKGROUND: Ethanol may be consumed for reasons such as reward, anxiety reduction, or caloric content, and the opioid enkephalin (ENK) appears to be involved in many of these functions. Previous studies in Sprague-Dawley rats have demonstrated that ENK in the hypothalamic paraventricular nucleus (PVN) is stimulated by voluntary consumption of ethanol. This suggests that this opioid peptide may be involved in promoting the drinking of ethanol, consistent with our recent findings that PVN injections of ENK analogs stimulate ethanol intake. To broaden our understanding of how this peptide functions throughout the brain to promote ethanol intake, we measured, in rats trained to drink 9% ethanol, the expression of the ENK gene in additional brain areas outside the hypothalamus, namely, the ventral tegmental area (VTA), nucleus accumbens shell (NAcSh) and core (NAcC), medial prefrontal cortex (mPFC), and central nucleus of the amygdala (CeA). METHODS: In the first experiment, the brains of rats chronically drinking 1 g/kg/d ethanol, 3 g/kg/d ethanol, or water were examined using real-time quantitative polymerase chain reaction (qRT-PCR). In the second experiment, a more detailed, anatomic analysis of changes in gene expression, in rats chronically drinking 3 g/kg/d ethanol compared to water, was performed using radiolabeled in situ hybridization (ISH). The third experiment employed digoxigenin-labeled ISH (DIG) to examine changes in the density of cells expressing ENK and, for comparison, dynorphin (DYN) in rats chronically drinking 3 g/kg/d ethanol versus water. RESULTS: With qRT-PCR, the rats chronically drinking ethanol plus water compared to water alone showed significantly higher levels of ENK mRNA, not only in the PVN but also in the VTA, NAcSh, NAcC, and mPFC, although not in the CeA. Using radiolabeled ISH, levels of ENK mRNA in rats drinking ethanol were found to be elevated in all areas examined, including the CeA. The experiment using DIG confirmed this effect of ethanol, showing an increase in density of ENK-expressing cells in all areas studied. It additionally revealed a similar change in DYN mRNA in the PVN, mPFC, and CeA, although not in the NAcSh or NAcC. CONCLUSIONS: While distinguishing the NAc as a site where ENK and DYN respond differentially, these findings lead us to propose that these opioids, in response to voluntary ethanol consumption, are generally elevated in extra-hypothalamic as well as hypothalamic areas, possibly to carry out specific area-related functions that, in turn, drive animals to further consume ethanol. These functions include calorie ingestion in the PVN, reward and motivation in the VTA and NAcSh, response-reinforcement learning in the NAcC, stress reduction in the CeA, and behavioral control in the mPFC.


Asunto(s)
Encefalinas/biosíntesis , Etanol/administración & dosificación , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/psicología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Dinorfinas/biosíntesis , Dinorfinas/fisiología , Encefalinas/fisiología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
11.
Neuropsychopharmacology ; 34(3): 775-85, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18800067

RESUMEN

Stress and anxiety are mainly regulated by amygdala and hypothalamic circuitries involving several neurotransmitter systems and providing physiological responses to peripheral organs via the hypothalamic-pituitary-adrenal axis and other pathways. The role of endogenous opioid peptides in this process is largely unknown. Here we show for the first time that anxiolytic parameters of explorative behavior in mice lacking prodynorphin were increased 2-4-fold in the open field, the elevated plus maze and the light-dark test. Consistent with this, treatment of wild-type mice with selective kappa-opioid receptor antagonists GNTI or norbinaltorphimine showed the same effects. Furthermore, treatment of prodynorphin knockout animals with U-50488H, a selective kappa-opioid receptor agonist, fully reversed their anxiolytic phenotype. These behavioral data are supported by an approximal 30% reduction in corticotropin-releasing hormone (CRH) mRNA expression in the hypothalamic paraventricular nucleus and central amygdala and an accompanying 30-40% decrease in corticosterone serum levels in prodynorphin knockout mice. Although stress-induced increases in corticosterone levels were attenuated in prodynorphin knockout mice, they were associated with minor increases in depression-like behavior in the tail suspension and forced swim tests. Taken together, our data suggest a pronounced impact of endogenous prodynorphin-derived peptides on anxiety, but not stress coping ability and that these effects are mediated via kappa-opioid receptors. The delay in the behavioral response to kappa-opioid receptor agonists and antagonist treatment suggests an indirect control level for the action of dynorphin, probably by modulating the expression of CRH or neuropeptide Y, and subsequently influencing behavior.


Asunto(s)
Ansiedad/metabolismo , Corticosterona , Encefalinas/fisiología , Neuropéptidos/metabolismo , Precursores de Proteínas/fisiología , Receptores Opioides kappa , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Tronco Encefálico/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/metabolismo , Dinorfinas/fisiología , Encefalinas/genética , Encefalinas/metabolismo , Conducta Exploratoria , Femenino , Guanidinas , Hipotálamo/metabolismo , Masculino , Aprendizaje por Laberinto , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/metabolismo , Morfinanos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuropéptido Y/fisiología , Neuropéptidos/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Núcleos del Rafe/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Estrés Psicológico/fisiopatología
12.
Neuroscience ; 144(1): 157-64, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17055175

RESUMEN

It has been shown that chronic cocaine increases prodynorphin mRNA in the caudate putamen and decreases it in the hypothalamus. In addition, treatment with a kappa-opioid receptor agonist produced the opposite effect on prodynorphin gene expression in these brain regions and also evoked a decrease in the hippocampus. It is already known that kappa-opioid receptor agonists decrease the development of sensitization to some of the behavioral effects of cocaine. The serotonin system has also been shown to regulate dynorphin gene expression and a continuous infusion of fluoxetine induced prodynorphin gene expression in the same pattern as the kappa-opioid agonist (+)(5a,7a,8b)-N-methyl-N-[7-(1-pyrrolidinyl)-1 oxaspiro[4.5]dec-8-yl]-benzeneacetamide (U-69593) in the brain regions investigated. It is interesting to note that treatment with a continuous infusion of cocaine produced different effects on this parameter. To determine whether serotonin plays a role in the regulation of prodynorphin mRNA by kappa-opioid agonists or cocaine, rats were treated with the serotonin depleter parachloroamphetamine (PCA). Beginning 24 h later, rats were treated with the selective kappa-opioid agonist U-69593 for 5 days or continuously with cocaine for 7 days and prodynorphin mRNA was measured. Prodynorphin mRNA was decreased significantly in the hypothalamus, caudate putamen, and hippocampus of rats treated with a single injection of PCA. Subsequent to PCA administration the effects of U-69593 or cocaine on prodynorphin mRNA were differentially affected across brain regions. Prodynorphin gene expression was still increased by U-69593 treatment in the hypothalamus and decreased in the caudate putamen. Cocaine treatment still produced a decrease in this parameter in the hypothalamus and an increase in the caudate putamen. In contrast, in the hippocampus, the decrease in prodynorphin mRNA produced by U-69593 was no longer evident after PCA and cocaine, which previously had no effect, now increased it in the serotonin-depleted group. These findings suggest that serotonin is necessary to maintain normal levels of dynorphin mRNA in all of the investigated brain areas and that the regulation of prodynorphin mRNA expression by chronic treatment with a kappa-opioid receptor agonist or cocaine requires serotonin in the hippocampus, but not in the hypothalamus or caudate putamen.


Asunto(s)
Sistema Nervioso Central/metabolismo , Cocaína/farmacología , Dinorfinas/fisiología , Receptores Opioides kappa/agonistas , Serotonina/fisiología , Animales , Bencenoacetamidas/farmacología , Northern Blotting , Sistema Nervioso Central/efectos de los fármacos , Encefalinas/biosíntesis , Fluoxetina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Precursores de Proteínas/biosíntesis , Pirrolidinas/farmacología , Sondas ARN , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
13.
J Neurosci ; 26(50): 13037-47, 2006 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-17167093

RESUMEN

The hypocretin/orexin arousal system plays a key role in maintaining an alert wake state. The hypocretin peptide is colocalized with an opioid peptide, dynorphin. As dynorphin may be coreleased with hypocretin, we asked what action simultaneous stimulation with the excitatory neuropeptide hypocretin and the inhibitory peptide dynorphin might exert on cells postsynaptic to hypocretin axons, including hypocretin neurons. Hypocretin neurons received direct synaptic contact from other hypocretin neurons but showed little direct response to hypocretin. Here, we show that mouse hypocretin neurons are acutely sensitive to dynorphin. Dynorphin inhibits the hypocretin system by direct postsynaptic actions (hyperpolarization, decreased spike frequency, increased GIRK (G-protein-gated inwardly rectifying K+ channel) current, and attenuated calcium current, and indirectly by reducing excitatory synaptic tone. Interestingly, a selective antagonist of kappa-opioid receptors enhanced activity of the hypocretin system, suggesting ongoing depression by endogenous hypothalamic opioids. Electrical stimulation of hypothalamic microslices that contained hypocretin cells and their axons evoked dynorphin release. Costimulation with dynorphin and hypocretin had three different effects on neurons postsynaptic to hypocretin axons: direct response to only one or the other of the two peptides [hypocretin cells respond to dynorphin, arcuate neuropeptide Y (NPY) cells respond to hypocretin], differential desensitization causing shift from inhibitory current to excitatory current with repeated coexposure (melanin-concentrating hormone neurons), synergistic direct excitation by hypocretin and presynaptic attenuation of inhibition by dynorphin (arcuate NPY neurons). These results suggest that hypocretin neurons may be able to exercise a high degree of modulatory control over postsynaptic targets using multiple neuropeptides with target-dependent actions.


Asunto(s)
Dinorfinas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/fisiología , Inhibición Neural/fisiología , Neuropéptidos/fisiología , Animales , Dinorfinas/biosíntesis , Dinorfinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Orexinas
14.
J Comp Neurol ; 493(1): 72-85, 2005 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-16255002

RESUMEN

We elaborate herein a novel theory of basal ganglia function that accounts for why palatable, energy-dense foods retain high incentive value even when immediate physiological energy requirements have been met. Basal ganglia function has been studied from the perspective of topographical segregation of processing within parallel circuits, with primary focus on motor control and cognition. Recent findings suggest, however, that the striatum can act as an integrated unit to modulate motivational state. We describe evidence that the striatal enkephalin system, which regulates the hedonic impact of preferred foods, undergoes coordinated gene expression changes that track current motivational state with regard to food intake. Striatal enkephalin gene expression is also downregulated by an intrastriatal infusion of a cholinergic muscarinic antagonist, a manipulation that greatly suppresses food intake. To account for these findings, we propose that signaling through a hypothalamic-midline thalamic-striatal axis impinges on the cholinergic interneurons of the striatum, which via their large, overlapping axonal fields act as a network to modulate enkephalin-containing striatal output neurons. A key relay in this circuit is the paraventricular thalamic nucleus, which receives convergent input from orexin-coded hypothalamic energy-sensing and behavioral state-regulating neurons, as well as from circadian oscillators, and projects to cholinergic interneurons throughout the striatal complex. We hypothesize that this system evolved to coordinate feeding and arousal, and to prolong the feeding central motivational state beyond the fulfillment of acute energy needs, thereby promoting "overeating" and the consequent development of an energy reserve for potential future food shortages.


Asunto(s)
Nivel de Alerta/fisiología , Metabolismo Energético/fisiología , Alimentos , Hipotálamo/fisiología , Recompensa , Tálamo/fisiología , Acetilcolina/farmacología , Acetilcolina/fisiología , Animales , Dopamina/farmacología , Dopamina/fisiología , Dinorfinas/farmacología , Dinorfinas/fisiología , Encefalinas/metabolismo , Encefalinas/fisiología , Hipotálamo/metabolismo , Modelos Biológicos , Narcóticos/metabolismo , Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiología , Precursores de Proteínas/metabolismo , Precursores de Proteínas/fisiología , ARN Mensajero/metabolismo , Ratas , Receptores Opioides kappa/metabolismo , Tálamo/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-15604626

RESUMEN

The concept of the placebo effect has received a considerable attention over the past several decades. The placebo effect has been observed in different psychiatric disorders, including post-traumatic stress disorder (PTSD), a chronic and severe disorder precipitated by exposure to a psychologically distressing event. The placebo response rates in patients with PTSD range from 19% to 62%. A considerable number of research publications suggest that endogenous opioids are involved in the mechanisms of the placebo effect. Endogenous opioid peptides play an important role in stress response and in the pathophysiology of PTSD. Therefore, endogenous opioids may be involved in the neurobiology of the placebo effect in PTSD. Possibly, the endogenous opioid system mediates the effect of placebo on all 3 PTSD symptom clusters (re-experiencing symptoms, avoidance and numbing, and physiologic arousal). The placebo effect-related activation of the endogenous opioid system may result in an improvement in intrusive symptomatology and symptoms of increased arousal because the administration of exogenous opioids improve these symptoms. The placebo effect-related activation of the endogenous opioid system may have a mood-enhancing effect, and, consequently, diminish avoidance and numbing. Multiple neurotransmitter and neuroendocrine pathways may be involved in the mechanisms of the placebo effect in PTSD. Further studies of the neurobiology of the placebo effect on patients with PTSD and other psychiatric disorders may produce interesting and important results.


Asunto(s)
Dinorfinas/fisiología , Endorfinas/fisiología , Encefalinas/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/fisiopatología , Humanos , Efecto Placebo , Trastornos por Estrés Postraumático/psicología , Estrés Psicológico/psicología
16.
Jpn J Pharmacol ; 79(3): 295-301, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10230857

RESUMEN

The analgesia-producing mechanism of processed Aconiti tuber was examined using rodents whose nociceptive threshold was decreased by loading repeated cold stress (RCS). The antinociceptive effect of processed Aconiti tuber (0.3 g/kg, p.o.) in RCS-loaded mice was antagonized by pretreatment with a kappa-opioid antagonist, nor-binaltorphimine (10 mg/kg, s.c.), and was abolished by an intrathecal injection of anti-dynorphin antiserum (5 microg). The Aconiti tuber-induced antinociception was inhibited by both dexamethasone (0.4 mg/kg, i.p.) and a dopamine D2 antagonist, sulpiride (10 mg/kg, i.p.), in RCS-loaded mice, and it was eliminated by both an electric lesion of the hypothalamic arcuate nucleus (HARN) and a highly selective dopamine D2 antagonist, eticlopride (0.05 microg), administered into the HARN in RCS-loaded rats. These results suggest that the analgesic effect of processed Aconiti tuber was produced via the stimulation of kappa-opioid receptors by dynorphin released in the spinal cord. It was also shown that dopamine D2 receptors in the HARN were involved in the expression of the analgesic activity of processed Aconiti tuber.


Asunto(s)
Analgésicos/farmacología , Medicamentos Herbarios Chinos/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Administración Tópica , Animales , Núcleo Arqueado del Hipotálamo/patología , Núcleo Arqueado del Hipotálamo/fisiopatología , Frío , Dexametasona/farmacología , Antagonistas de Dopamina/farmacología , Dinorfinas/inmunología , Dinorfinas/fisiología , Glucocorticoides/farmacología , Hipotálamo/patología , Hipotálamo/fisiopatología , Sueros Inmunes/inmunología , Sueros Inmunes/farmacología , Ligandos , Masculino , Ratones , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Nociceptores/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides kappa/fisiología , Receptores Opioides mu/antagonistas & inhibidores , Salicilamidas/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Sulpirida/farmacología
17.
Brain Res ; 732(1-2): 113-20, 1996 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-8891275

RESUMEN

The purpose of the present study was to determine if D2 receptor-mediated activation of hypothalamic tuberoinfundibular dopaminergic (TIDA) neurons occurs via afferent neuronal inhibition of tonically active inhibitory dynorphinergic neurons in the male rat. To this end, the effects of either surgical deafferentation of the mediobasal hypothalamus or administration of a kappa opioid receptor agonist (U-50,488) or antagonist (nor-binaltorphimine (NOR-BNI)) on D2 receptor-mediated activation of TIDA neurons were assessed. For comparison, the activity of mesolimbic DA neurons was also determined in these studies. TIDA and mesolimbic DA neuronal activities were estimated by measuring dopamine synthesis (accumulation of 3,4-dihydroxyphenylalanine (DOPA) following decarboxylase inhibition) and metabolism (concentrations of 3,4-dihydroxyphenylacetic acid (DOPAC)) in terminals of these neurons in the median eminence and nucleus accumbens, respectively. Intraperitoneal administration of the D2 receptor agonist quinelorane caused a dose-dependent increase in DOPAC in the median eminence and a decrease in DOPAC in the nucleus accumbens; surgical deafferentation of the mediobasal hypothalamus prevented the effect of quinelorane in the median eminence, but not the nucleus accumbens. Activation of kappa opioid receptors with U-50,488 had no effect per se, but blocked quinelorane-induced increases in median eminence DOPA. In contrast, U-50,488 had no effect on DOPA in the nucleus accumbens of either vehicle- or quinelorane-treated rats. Blockade of kappa opioid receptors with NOR-BNI increased median eminence DOPA, and prevented the stimulatory effects of quinelorane on dopamine synthesis. Administration of prolactin also increased median eminence DOPA, but did not alter the ability of quinelorane to stimulate dopamine synthesis. Neither NOR-BNI nor prolactin had any effect on DOPA in the nucleus accumbens of vehicle- or quinelorane-treated rats. These results suggest that D2 receptor-mediated activation of TIDA neurons occurs via an afferent neuronal mechanism involving, at least in part, inhibition of tonically active inhibitory dynorphinergic neurons in the male rat.


Asunto(s)
Dopamina/metabolismo , Dinorfinas/fisiología , Hipotálamo Medio/fisiología , Hipotálamo/fisiología , Naltrexona/análogos & derivados , Neuronas/fisiología , Pirrolidinas/farmacología , Receptores de Dopamina D2/fisiología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero , Ácido 3,4-Dihidroxifenilacético/metabolismo , Vías Aferentes/fisiología , Analgésicos/administración & dosificación , Analgésicos/farmacología , Animales , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/fisiología , Dihidroxifenilalanina/metabolismo , Dopa-Decarboxilasa/metabolismo , Agonistas de Dopamina/administración & dosificación , Agonistas de Dopamina/farmacología , Hipotálamo/efectos de los fármacos , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Sistema Límbico/citología , Sistema Límbico/fisiología , Masculino , Eminencia Media/fisiología , Naltrexona/administración & dosificación , Naltrexona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Núcleo Accumbens/fisiología , Prolactina/administración & dosificación , Prolactina/farmacología , Pirrolidinas/administración & dosificación , Quinolinas/administración & dosificación , Quinolinas/farmacología , Ratas , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores
18.
Behav Brain Res ; 47(2): 143-9, 1992 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-1350448

RESUMEN

The cross-tolerance technique was used to analyze the receptor mechanisms of analgesia induced by electroacupuncture (EA) of 2 Hz, 100 Hz, or 2-15 Hz. (1) Rats were given EA stimulation of 2 Hz, 100 Hz and 2-15 Hz for 30 min with 30 min intervals successively. The percentage increase in tail-flick latency (TFL) was taken to indicate the intensity of EA analgesia. Rats made tolerant to repeated intrathecal injection of the mu-opioid agonist ohmefentanyl (OMF, 15 pmol, Q2h x 5) or the delta-opioid agonist DPDPE (10 nmol, Q2h x 5) showed a cross tolerance to both 2 Hz- and 2-15 Hz-, but not to 100 Hz-EA analgesia; and rats made tolerant to kappa-opioid agonist dynorphin-(1-13) (5 nmol, Q2h x 5) showed a cross-tolerance to 100 Hz- and 2-15 Hz-, but not to 2 Hz-EA analgesia; (2) Rats made tolerant to 2-15 Hz EA showed cross-tolerance to either 2 Hz- or 100 Hz-EA analgesia; (3) Rats made tolerant to either 2 Hz- or 100 Hz-EA were still reactive to 2-15 Hz-EA. The results indicate that 2 Hz-EA analgesia is mediated by mu- and delta-receptors, 100 Hz-EA analgesia by kappa-receptor, and 2-15 Hz-EA analgesia by combined action of mu-, delta- and kappa-receptors in the spinal cord of the rats.


Asunto(s)
Electroacupuntura , Endorfinas/fisiología , Nociceptores/fisiología , Receptores Opioides/fisiología , Animales , Dinorfinas/fisiología , Ratas , Ratas Endogámicas , Tiempo de Reacción/fisiología , Receptores Opioides delta , Receptores Opioides kappa , Receptores Opioides mu , Umbral Sensorial/fisiología
19.
Brain Res ; 374(2): 236-43, 1986 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-2872940

RESUMEN

Leumorphin, an opioid peptide whose functions are unknown, is found in mammalian brain and pituitary and stimulates lordosis behavior in estrogen-treated female rats. To elucidate the role of leumorphin in the physiological control of female sexual behavior, the levels of immunoreactive (ir) leumorphin as well as ir dynorphin (dynorphin A) were measured in the rat brain and pituitary during the estrous cycle. There was a clear variation of ir leumorphin in the hypothalamus and anterior pituitary during the estrous cycle. The levels of ir leumorphin in the hypothalamus and anterior pituitary on the afternoon of proestrus were significantly higher (P less than 0.01) than those on the afternoons of estrus and metestrus. The rise in the hypothalamic levels of ir leumorphin on the afternoon of proestrus was correlated with the receptivity of lordosis during the estrous cycle. Furthermore, there was a close correlation with ir dynorphin levels. These findings are in agreement with studies demonstrating a common precursor for leumorphin and dynorphin. Ir leumorphin in the hippocampus and neurointermediate pituitary did not change significantly during the estrous cycle. Because the leumorphin antiserum used recognizes rimorphin (dynorphin B) 1.78 times more than porcine leumorphin on a molar basis, high performance-gel permeation chromatography was done on pooled extracts of hypothalamus taken at proestrus and estrus. The peak in the leumorphin-like substance in the activation of sexual behavior is discussed.


Asunto(s)
Encefalinas/metabolismo , Estro , Hipotálamo/metabolismo , Adenohipófisis/metabolismo , Precursores de Proteínas/metabolismo , Conducta Sexual Animal , Animales , Reacciones Antígeno-Anticuerpo , Dinorfinas/aislamiento & purificación , Dinorfinas/metabolismo , Dinorfinas/fisiología , Encefalinas/aislamiento & purificación , Encefalinas/fisiología , Estro/efectos de los fármacos , Femenino , Hipotálamo/fisiología , Adenohipófisis/fisiología , Precursores de Proteínas/aislamiento & purificación , Precursores de Proteínas/fisiología , Ratas , Ratas Endogámicas , Conducta Sexual Animal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA