Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Neuroendocrinology ; 103(6): 787-805, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26765570

RESUMEN

Estradiol rapidly regulates the activity of arcuate nucleus (ARH) proopiomelanocortin (POMC) neurons that project to the medial preoptic nucleus (MPN) to regulate lordosis. Orphanin FQ/nociceptin (OFQ/N) acts via opioid receptor-like (ORL)-1 receptors to inhibit these POMC neurons. Therefore, we tested the hypothesis that estradiol excites POMC neurons by rapidly attenuating inhibitory ORL-1 signaling in these cells. Hypothalamic slices through the ARH were prepared from ovariectomized rats injected with Fluorogold into the MPN. Electrophysiological recordings were generated in ARH neurons held at or near -60 mV, and neuronal phenotype was determined post hoc by immunohistofluorescence. OFQ/N application induced robust outward currents and hyperpolarizations via G protein-gated, inwardly rectifying K+ (GIRK) channels that were attenuated by pretreatment with either 17-ß estradiol (E2) or E2 conjugated to bovine serum albumin. This was blocked by the estrogen receptor (ER) antagonist ICI 182,780 and mimicked by the Gq-coupled membrane ER (Gq-mER) ligand STX and the ERα agonist PPT. Inhibiting phosphatidylinositol-3-kinase (PI3K) blocked the estrogenic attenuation of ORL-1/GIRK currents. Antagonizing either phospholipase C (PLC), protein kinase C (PKC), protein kinase A (PKA) or neuronal nitric oxide synthase (nNOS) also abrogated E2 inhibition of ORL-1/GIRK currents, whereas activation of PKC, PKA, protein kinase B (Akt) and nNOS substrate L-arginine all attenuated the OFQ/N response. This was observed in 92 MPN-projecting, POMC-positive ARH neurons. Thus, ORL-1 receptor-mediated inhibition of POMC neurons is rapidly and negatively modulated by E2, an effect which is stereoselective and membrane initiated via Gq-mER and ERα activation that signals through PLC, PKC, PKA, PI3K and nNOS.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptores Opioides/metabolismo , Animales , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Femenino , Hipotálamo/citología , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Péptidos Opioides/farmacología , Ovariectomía , Piperidinas/farmacología , Pirrolidinonas/farmacología , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Estilbamidinas/farmacocinética , Tetrodotoxina/farmacología , Receptor de Nociceptina , Nociceptina
2.
Pharmacol Rep ; 67(5): 1010-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26398397

RESUMEN

BACKGROUND: Kahweol is a diterpene present in the oil derived from coffee beans. Although several pharmacological activities of kahweol are already well described in the literature, no study was done in order to assess the analgesic activity of this substance. Thus, the aim of this study was to investigate the possible peripheral antinociceptive effect of kahweol. Considering that the opioid peptides have been implicated in peripheral antinociception induced by non-opioidergic compounds, the present study also evaluated the endogenous opioids involvement in this effect. METHODS: The rat paw pressure test was used, and hyperalgesia was induced by intraplantar injection of prostaglandin E2 (2µg/paw). All drugs were administered subcutaneously in the hindpaws of male Wistar rats. The expression of ß-endorphin was examined by immunohistochemistry in the skin tissue samples of the plantar surface of rat right hindpaws. RESULTS: Intraplantar injection of kahweol (40 and 80µg) induced significant peripheral antinociception. The antinociceptive effect of kahweol was due to a local peripheral action because the higher dose (80µg/paw) did not produce any effect in the contralateral paw. The opioid receptor antagonist naloxone (50 and 100µg/paw) prevented action of kahweol (80µg/paw) and the aminopeptidases inhibitor bestatin (400µg/paw) potentiated the antinociceptive effect of kahweol (40µg/paw). Furthermore, kahweol treatment increased the intensity of ß-endorphin immunoreactivity in the epithelium of rat paws. CONCLUSIONS: The results discussed here provide evidence that kahweol treatment has peripheral antinociceptive effect and suggest that this effect is mediated by the release of endogenous opioids.


Asunto(s)
Analgésicos/farmacología , Café/química , Diterpenos/farmacología , Péptidos Opioides/farmacología , Animales , Dinoprostona , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Dimensión del Dolor , Péptidos/farmacología , Presión , Ratas , Ratas Wistar , Piel/efectos de los fármacos , Piel/metabolismo , betaendorfina/biosíntesis
3.
Neuropeptides ; 50: 51-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25812480

RESUMEN

Nociceptin/Orphanin FQ (N/OFQ) is a 17-amino acid peptide that binds to the nociceptin receptor (NOP). N/OFQ and NOP receptors are expressed in numerous brain areas. The generation of specific agonists, antagonists and receptor-deficient mice or rats has enabled progress in elucidating the biological functions of N/OFQ. These tools have been employed to identify the biological significance of the N/OFQ system and how it interacts with other endogenous systems to regulate several body functions. The present review focuses on the role of N/OFQ in the regulation of body temperature and its relationship with energy balance. Critical evaluation of the literature data suggests that N/OFQ, acting through the NOP receptor, may cause hypothermia by influencing the complex thermoregulatory system that operates as a federation of independent thermoeffector loops to control body temperature at the hypothalamic level. Furthermore, N/OFQ counteracts hyperthermia elicited by cannabinoids or µ-opioid agonists. N/OFQ-induced hypothermia is prevented by ω-conotoxin GVIA, an N-type calcium channel blocker. Hypothermia induced by N/OFQ is considered within the framework of the complex action that this neuropeptide exerts on energy balance. Energy stores are regulated through the complex neural controls exerted on both food intake and energy expenditure. In laboratory rodents, N/OFQ stimulates consummatory behavior and decreases energy expenditure. Taken together, these studies support the idea that N/OFQ contributes to the regulation of energy balance by acting as an "anabolic" neuropeptide as it elicits effects similar to those produced in the hypothalamus by other neuropeptides such as orexins and neuropeptide Y.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Hipotálamo/fisiología , Péptidos Opioides/fisiología , Animales , Química Encefálica , Citocinas/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Fiebre/fisiopatología , Hiperfagia/fisiopatología , Ratones , Neuropéptidos/fisiología , Obesidad/fisiopatología , Péptidos Opioides/genética , Péptidos Opioides/farmacología , ARN Mensajero/análisis , Ratas , Receptores Opioides/genética , Receptores Opioides/fisiología , Aumento de Peso/fisiología , Receptor de Nociceptina , Nociceptina
4.
Mol Pain ; 10: 10, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24499354

RESUMEN

BACKGROUND: Leukocytes containing opioid peptides locally control inflammatory pain. In the early phase of complete Freund's adjuvant (CFA)-induced hind paw inflammation, formyl peptides (derived e.g. from Mycobacterium butyricum) trigger the release of opioid peptides from neutrophils contributing to tonic basal antinociception. In the later phase we hypothesized that toll-like-receptor-(TLR)-4 activation of monocytes/macrophages triggers opioid peptide release and thereby stimulates peripheral opioid-dependent antinociception. RESULTS: In Wistar rats with CFA hind paw inflammation in the later inflammatory phase (48-96 h) systemic leukocyte depletion by cyclophosphamide (CTX) or locally injected naloxone (NLX) further decreased mechanical and thermal nociceptive thresholds. In vitro ß-endorphin (ß-END) content increased during human monocyte differentiation as well as in anti-inflammatory CD14+CD16- or non-classical M2 macrophages. Monocytes expressing TLR4 dose-dependently released ß-END after stimulation with lipopolysaccharide (LPS) dependent on intracellular calcium. Despite TLR4 expression proinflammatory M1 and anti-inflammatory M2 macrophages only secreted opioid peptides in response to ionomycin, a calcium ionophore. Intraplantar injection of LPS as a TLR4 agonist into the inflamed paw elicited an immediate opioid- and dose-dependent antinociception, which was blocked by TAK-242, a small-molecule inhibitor of TLR4, or by peripheral applied NLX. In the later phase LPS lowered mechanical and thermal nociceptive thresholds. Furthermore, local peripheral TLR4 blockade worsened thermal and mechanical nociceptive pain thresholds in CFA inflammation. CONCLUSION: Endogenous opioids from monocytes/macrophages mediate endogenous antinociception in the late phase of inflammation. Peripheral TLR4 stimulation acts as a transient counter-regulatory mechanism for inflammatory pain in vivo, and increases the release of opioid peptides from monocytes in vitro. TLR4 antagonists as new treatments for sepsis and neuropathic pain might unexpectedly transiently enhance pain by impairing peripheral opioid analgesia.


Asunto(s)
Analgesia , Inflamación/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Receptor Toll-Like 4/metabolismo , Animales , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/farmacología , Humanos , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Hiperalgesia/patología , Inflamación/complicaciones , Inflamación/metabolismo , Inflamación/patología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Nocicepción/efectos de los fármacos , Péptidos Opioides/farmacología , Ratas , Ratas Wistar , Receptores de IgG/metabolismo , Receptores Opioides/metabolismo , Receptor Toll-Like 2/metabolismo , betaendorfina/metabolismo
5.
J Nutr Biochem ; 25(5): 503-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24411973

RESUMEN

Human milk contains a multitude of bioactive proteins with very diverse functions, which are beneficial for the rapidly growing neonate. The large variety of bioactivities is accomplished by the combination of bioactive proteins per se and gastrointestinal release of bioactive peptides derived from them. The bioactivities exerted by these peptides include enhancement of mineral absorption, immunomodulation, opioid, antihypertensive and antimicrobial activities. Notably, several of the activities are not attributed to the parental proteins, but exclusively to released bioactive peptides. This article reviews studies on bioactive peptides derived from major human milk proteins, such as caseins, α-lactalbumin and lactoferrin, during gastrointestinal digestion. Studies of bovine milk counterparts are also cited as a comparison.


Asunto(s)
Proteínas de la Leche/química , Péptidos/farmacología , Adyuvantes Inmunológicos/farmacología , Secuencia de Aminoácidos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antioxidantes/farmacología , Caseínas/farmacología , Bovinos , Humanos , Absorción Intestinal , Lactoferrina/farmacología , Datos de Secuencia Molecular , Péptidos Opioides/farmacología , Fragmentos de Péptidos/farmacología , Fosfopéptidos/farmacología
6.
Endocrinology ; 154(11): 4249-58, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23928375

RESUMEN

Orphanin FQ (OFQ), a member of the opioid family, is found in many areas of the hypothalamus and, when given centrally OFQ inhibits episodic LH secretion in rodents and sheep. Because GnRH neurons are devoid of the appropriate receptors to mediate steroid negative feedback directly, neurons that release OFQ may be involved. Using immunocytochemistry, we first determined that most OFQ neurons in the arcuate nucleus (ARC) and other hypothalamic regions of luteal phase ewes contained both estrogen receptor α and progesterone (P) receptor. Given a similar high degree of steroid receptor colocalization in other ARC subpopulations, we examined whether OFQ neurons of the ARC contained those other neuropeptides and neurotransmitters. OFQ did not colocalize with kisspeptin, tyrosine hydroxylase, or agouti-related peptide, but all ARC OFQ neurons coexpressed proopiomelanocortin. To test for a role for endogenous OFQ, we examined the effects of an OFQ receptor antagonist, [Nphe1,Arg14,Lys15]Nociceptin-NH2 (UFP-101) (30 nmol intracerebroventricular/h), on LH secretion in steroid-treated ewes in the breeding season and ovary-intact ewes in anestrus. Ovariectomized ewes with luteal phase concentrations of P and estradiol showed a significant increase in LH pulse frequency during infusion of UFP-101 (4.5 ± 0.5 pulses/6 h) compared with saline infusion (2.6 ± 0.4 pulses/6 h), whereas ewes implanted with only estradiol did not. Ovary-intact anestrous ewes displayed no significant differences in LH pulse amplitude or frequency during infusion of UFP-101. Therefore, we conclude that OFQ mediates, at least in part, the negative feedback action of P on GnRH/LH pulse frequency in sheep.


Asunto(s)
Retroalimentación Fisiológica , Péptidos Opioides/metabolismo , Progesterona/metabolismo , Ovinos/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/administración & dosificación , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estro , Femenino , Hipotálamo/metabolismo , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Neuronas/metabolismo , Péptidos Opioides/administración & dosificación , Péptidos Opioides/genética , Péptidos Opioides/farmacología , Ovariectomía , Progesterona/administración & dosificación , Progesterona/farmacología , Transporte de Proteínas , Receptores de Progesterona/metabolismo , Nociceptina
7.
Psychopharmacology (Berl) ; 228(1): 53-63, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23455592

RESUMEN

RATIONALE: Nociceptin/orphanin FQ (N/OFQ) is a functional antagonist of corticotrophin-releasing factor, the main mediator of the stress response. Stress represents a key determinant of binge eating (BE) for highly palatable food (HPF). OBJECTIVES: In relation to the antistress properties of N/OFQ, we evaluated its effect on BE. After the observation that episodes of food restriction increase the sensitivity to its hyperphagic effects, the function of NOP receptor and N/OFQ was investigated after cycles of food restrictions. MATERIALS AND METHODS: In BE experiments, four groups were used: rats fed normally and not stressed or stressed, rats exposed to cycles of restriction/refeeding and then stressed, or not stressed. In the other experiments, two groups were used: rats exposed or not to food restriction. RESULTS: Only restricted and stressed rats exhibited BE for HPF (containing chocolate cream). Intracerebroventricular injections of N/OFQ of 0.5 nmol/rat significantly reduced BE. N/OFQ 1 nmol/rat did not reduce BE but significantly increased HPF intake following food restrictions. Cycles of food restriction increased animals' sensitivity to the hyperphagic effect of N/OFQ for HPF. In situ hybridization studies following food restrictions showed decreased ppN/OFQ mRNA expression in the bed nucleus of the stria terminalis and increased expression of ppN/OFQ and NOP receptor mRNA in the ventral tegmental area and in the ventromedial hypothalamus, respectively. CONCLUSIONS: These findings indicate that N/OFQ slightly reduces BE at low doses, while higher doses increase HPF intake, due to increased sensitivity to its hyperphagic effect following a history of caloric restrictions.


Asunto(s)
Bulimia/prevención & control , Restricción Calórica , Péptidos Opioides/farmacología , Estrés Psicológico/tratamiento farmacológico , Animales , Bulimia/etiología , Relación Dosis-Respuesta a Droga , Femenino , Hiperfagia/etiología , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Péptidos Opioides/administración & dosificación , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/genética , Receptores Opioides/metabolismo , Estrés Fisiológico/efectos de los fármacos , Estrés Psicológico/complicaciones , Área Tegmental Ventral/metabolismo , Receptor de Nociceptina , Nociceptina
8.
Horm Behav ; 60(5): 540-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21872598

RESUMEN

Sexual receptivity, lordosis, can be induced by sequential estradiol and progesterone or extended exposure to high levels of estradiol in the female rat. In both cases estradiol initially inhibits lordosis through activation of ß-endorphin (ß-END) neurons of the arcuate nucleus of the hypothalamus (ARH) that activate µ-opioid receptors (MOP) in the medial preoptic nucleus (MPN). Subsequent progesterone or extended estradiol exposure deactivates MPN MOP to facilitate lordosis. Opioid receptor-like receptor-1 (ORL-1) is expressed in ARH and ventromedial hypothalamus (VMH). Infusions of its endogenous ligand, orphanin FQ (OFQ/N, aka nociceptin), into VMH-ARH region facilitate lordosis. Whether OFQ/N acts in ARH and/or VMH and whether OFQ/N is necessary for steroid facilitation of lordosis are unclear. In Exp I, OFQ/N infusions in VMH and ARH that facilitated lordosis also deactivated MPN MOP indicating that OFQ/N facilitation of lordosis requires deactivation of ascending ARH-MPN projections by directly inhibiting ARH ß-END neurons and/or through inhibition of excitatory VMH-ARH pathways to proopiomelanocortin neurons. It is unclear whether OFQ/N activates the VMH output motor pathways directly or via the deactivation of MPN MOP. In Exp II we tested whether ORL-1 activation is necessary for estradiol-only or estradiol+progesterone lordosis facilitation. Blocking ORL-1 with UFP-101 inhibited estradiol-only lordosis and MPN MOP deactivation but had no effect on estradiol+progesterone facilitation of lordosis and MOP deactivation. In conclusion, steroid facilitation of lordosis inhibits ARH ß-END neurons to deactivate MPN MOP, but estradiol-only and estradiol+progesterone treatments appear to use different neurotransmitter systems to inhibit ARH-MPN signaling.


Asunto(s)
Hipotálamo/efectos de los fármacos , Péptidos Opioides/fisiología , Postura/fisiología , Conducta Sexual Animal/fisiología , Animales , Estradiol/administración & dosificación , Femenino , Hipotálamo/fisiología , Masculino , Péptidos Opioides/administración & dosificación , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/farmacología , Área Preóptica , Progesterona/administración & dosificación , Ratas , Ratas Long-Evans , Receptores Opioides mu/fisiología , Conducta Sexual Animal/efectos de los fármacos , Nociceptina
9.
Neuropharmacology ; 59(3): 190-200, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20510254

RESUMEN

We evaluated the role of the nociceptin/orphanin FQ (NOP) receptor in regulating food intake, meal pattern and the activity of hypothalamic arcuate (ARC) neurons. The microstructural analysis of food intake and meal pattern was performed under both food-deprived and ad libitum conditions. Whole-cell patch clamp recordings were obtained using the in vitro hypothalamic slice preparation and biocytin-filled electrodes. NOP receptor knockout mice exhibited significantly reduced body weight. Fasting-induced hyperphagia was diminished for the first 2h of a 6-h re-feeding period, and was associated with decreased meal duration and size, as well as a biphasic effect on meal frequency. The genotype effect observed under ad libitum conditions was comparatively unremarkable. Orphanin FQ/nociceptin (OFQ/N) was able to decrease evoked excitatory postsynaptic current amplitude, increase the S(2):S(1) ratio via the paired-pulse paradigm, and decrease miniature excitatory postsynaptic current frequency in ARC neurons from wild type animals but not NOP receptor knockouts. In addition OFQ/N activated a reversible outward current that was antagonized by the G-protein activated, inwardly-rectifying K(+) (GIRK) channel blocker tertiapin in wild type but not NOP knockout animals. Both the presynaptic and postsynaptic actions of OFQ/N were observed in ARC neurons subsequently determined to be immunopositive for characteristic phenotypic markers of anorexigenic proopiomelanocortin (POMC) neurons. Taken together, these results demonstrate the contribution of the NOP receptor in controlling food intake and meal pattern, as well as glutamate release and GIRK1 channel activity at POMC synapses.


Asunto(s)
Regulación del Apetito/genética , Potenciales Postsinápticos Excitadores/genética , Conducta Alimentaria/fisiología , Neuronas/fisiología , Proopiomelanocortina/metabolismo , Receptores Opioides/metabolismo , Análisis de Varianza , Animales , Regulación del Apetito/efectos de los fármacos , Biofisica , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Hipotálamo/citología , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Péptidos Opioides/farmacología , Técnicas de Placa-Clamp/métodos , Receptores Opioides/deficiencia , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología , Receptor de Nociceptina , Nociceptina
10.
Mol Pharmacol ; 77(5): 804-17, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159949

RESUMEN

The pharmacology of G protein-coupled receptors can be influenced by factors such as constitutive receptor activation and Na(+) ions. In this study, we examined the coupling of natively and heterologously expressed nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptors with voltage-dependent Ca(2+) channels after exposure to four high-affinity NOP receptor blockers [[Nphe(1)Arg(14)Lys(15)]N/OFQ-NH(2) (UFP-101), 1-[1-(cyclooctylmethyl)-1,2,3,6-tetrahydro-5-(hydroxymethyl)-4-pyridinyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one (Trap-101), 1-benzyl-N-{3-[spiroisobenzofuran-1(3H),4'-piperidin-1-yl]propyl}pyrrolidine-2-carboxamide (compound 24), and N-(4-amino-2-methylquinolin-6-yl)-2-(4-ethylphenoxymethyl)benzamide hydrochloride (JTC-801)] in sympathetic neurons. The enhanced tonic inhibition of Ca(2+) currents in the absence of agonists, indicative of constitutively active NOP receptors in transfected neurons, was abolished after pretreatment with pertussis toxin. In control neurons, the four antagonists did not exert any effects when applied alone but significantly blocked the N/OFQ-mediated Ca(2+) current inhibition. Exposure of transfected neurons to UFP-101 resulted in partial agonist effects. In contrast, Trap-101, compound 24, and JTC-801 exerted inverse agonism, as measured by the loss of tonic Ca(2+) current inhibition. In experiments designed to measure the N/OFQ concentration-response relationship under varying Na(+) concentrations, a leftward shift of IC(50) values was observed after Na(+) exposure. Although similar N/OFQ efficacies were measured with all solutions, a significant decrease of Hill coefficient values was obtained with increasing Na(+) concentrations. Examination of the allosteric effects of Na(+) on heterologously overexpressed NOP receptors showed that the tonic Ca(2+) current inhibition was abolished in the presence of the monovalent cation. These results demonstrate that constitutively active NOP receptors exhibit differential blocker pharmacology and allosteric regulation by Na(+). Data are also presented demonstrating that heterologously expressed mu opioid receptors in sympathetic neurons are similarly modulated.


Asunto(s)
Neuronas/fisiología , Péptidos Opioides/farmacología , Receptores Opioides/genética , Sodio/farmacología , Sistema Nervioso Simpático/fisiología , Animales , Calcio/farmacología , Calcio/fisiología , ADN Complementario/genética , Electrofisiología/métodos , Antagonistas de Narcóticos , Neuronas/efectos de los fármacos , Plásmidos , Ratas , Receptores Opioides/agonistas , Receptores Opioides/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Transfección , Receptor de Nociceptina
11.
Neuroscience ; 166(1): 40-8, 2010 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-20006677

RESUMEN

The contribution of endogenous nociceptin/orphanin FQ (N/OFQ) to neuroleptic-induced parkinsonism has been evaluated in haloperidol-treated mice. Pharmacological blockade of N/OFQ receptors (NOP) via systemic administration of 1-[(3R,4R)-1-cyclooctylmethyl-3-hydroxymethyl-4-piperidyl]-3-ethyl-1,3-dihydro-2H benzimidazol-2-one (J-113397, 0.01-10 mg/kg i.p.) or central injection of [Nphe(1),Arg(14),Lys(15)]N/OFQ-NH(2) (UFP-101, 10 nmol i.c.v.) attenuated (0.8 mg/kg) haloperidol-induced motor deficits as evaluated by a battery of behavioral tests providing complementary information on motor parameters: the bar, drag and rotarod tests. A combined neurochemical and behavioral approach was then used to investigate whether the substantia nigra reticulata could be involved in antiakinetic actions of J-113397. Microdialysis combined to the bar test revealed that haloperidol (0.3 and 0.8 mg/kg i.p.) caused a dose-dependent and prolonged elevation of immobility time (i.e. akinesia) which was associated with an increase in nigral glutamate and a reduction in GABA release. Conversely, J-113397 (1 mg/kg) alone reduced glutamate and elevated nigral GABA release, and when challenged against haloperidol, counteracted its behavioral and neurochemical effects. Microdialysis coupled to behavioral testing also demonstrated that NOP receptor knockout mice were resistant to haloperidol (0.3 mg/kg) compared to wild-type mice, lack of response being associated with a reversal of glutamate release facilitation into inhibition and no change in nigral GABA release. This study provides pharmacological and genetic evidence that endogenous N/OFQ contributes to haloperidol-induced akinesia and changes of amino acid transmission in mice. Moreover, it confirms the view that NOP receptor antagonists are capable of reversing akinesia across species and genotypes and may prove effective in relieving neuroleptic-induced parkinsonism.


Asunto(s)
Discinesia Inducida por Medicamentos/metabolismo , Haloperidol/farmacología , Antagonistas de Narcóticos , Trastornos Parkinsonianos/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Animales , Antipsicóticos/farmacología , Bencimidazoles/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/fisiopatología , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Ácido Glutámico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Neurofarmacología/métodos , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/fisiopatología , Piperidinas/farmacología , Receptores Opioides/genética , Sustancia Negra/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Receptor de Nociceptina , Nociceptina
12.
Am J Physiol Heart Circ Physiol ; 297(2): H859-65, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19561314

RESUMEN

The role of nociceptin and its spinal cord neural pathways in electroacupuncture (EA)-related inhibition of visceral excitatory reflexes is not clear. Nociceptin/orphanin FQ (N/OFQ) is an endogenous ligand for a G protein-coupled receptor, called the N/OFQ peptide (NOP) receptor, which has been found to be distributed in the spinal cord. The present study investigated the importance of this system in visceral-cardiovascular reflex modulation during EA. Cardiovascular pressor reflex responses were induced by gastric distension in Sprague-Dawley rats anesthetized by ketamine and xylazine. An intrathecal injection of nociceptin (10 nM) at T1-2 attenuated the pressor responses by 35%, similar to the influence of EA at P 5-6 (42% decrease). An intrathecal injection of the NOP antagonist, [N-Phe(1)]nociceptin(1-13) NH(2), partially reversed the EA response. Pretreatment with the opioid receptor antagonist naloxone did not alter the EA-like inhibitory effect of nociceptin on the pressor reflex, whereas a combination of nociceptin receptor antagonist with naloxone completely abolished the EA response. An intrathecal injection of nociceptin attenuated the pressor responses to the electrical stimulation of the rostral ventrolateral medulla by 46%, suggesting that nociceptin can regulate sympathetic outflow. Furthermore, a bilateral microinjection of NOP antagonist into either the dorsal horn or the intermediolateral column at T1 partially reversed the EA inhibitory effect. These results suggest that nociceptin in the spinal cord mediates part of the EA-related modulation of visceral reflex responses.


Asunto(s)
Electroacupuntura , Péptidos Opioides/fisiología , Células del Asta Posterior/fisiología , Reflejo/fisiología , Sistema Nervioso Simpático/fisiología , Animales , Estimulación Eléctrica , Vaciamiento Gástrico/efectos de los fármacos , Vaciamiento Gástrico/fisiología , Inyecciones Espinales , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/antagonistas & inhibidores , Péptidos Opioides/farmacología , Células del Asta Posterior/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Estómago/inervación , Estómago/fisiología , Vértebras Torácicas , Aferentes Viscerales/fisiología , Nociceptina
13.
Neurosci Lett ; 456(2): 59-63, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19429134

RESUMEN

Activation of opioid receptor-like 1 receptor (ORL(1)) by intrathecal administration of orphanin FQ (OFQ), an endogenous ligand for the ORL(1) receptor, has been shown to produce antinociception. In addition, we have recently shown gonadal hormone-dependent, sex-specific modulation of acute spinal nociception such that estrogen attenuated OFQ-induced antinociception in the female whereas testosterone was required for the expression of antinociception in the male. However, sex-related differences in the role of OFQ under hyperalgesic conditions are unknown. Hence, we investigated whether OFQ produces sex-specific modulation of mustard oil-induced secondary thermal hyperalgesia in the rat. Mustard oil application to the hind limb significantly reduced the tail-flick latencies (TFL) in male, and ovariectomized (OVX), estradiol treated ovariectomized (OVX+E), proestrous (ProE) and diestrous (DiE) females. Intrathecal administration of OFQ not only attenuated mustard oil-induced decrease in TFLs, i.e. reversed hyperalgesia, but also led to a significant increase in TFLs above the baseline, i.e. produced antinociception in male, OVX, and diestrous rats. However, OFQ failed to alter TFLs in proestrous or OVX+E females, thus these two groups with elevated estrogen levels remained hyperalgesic following mustard oil treatment. These findings demonstrate that OFQ modulates mustard oil-induced secondary hyperalgesia in an estrogen-dependent, sex-specific manner.


Asunto(s)
Estrógenos/metabolismo , Hiperalgesia/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Caracteres Sexuales , Animales , Femenino , Calor , Hiperalgesia/inducido químicamente , Irritantes/toxicidad , Masculino , Planta de la Mostaza/toxicidad , Péptidos Opioides/farmacología , Ovariectomía , Umbral del Dolor/efectos de los fármacos , Aceites de Plantas/toxicidad , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptor de Nociceptina , Nociceptina
14.
Gen Comp Endocrinol ; 163(1-2): 47-51, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19318106

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) was identified in 1995 as the endogenous ligand for the orphan G(i)/G(o)-coupled opioid receptor-like 1 receptor (NOP(1)). Exogenous N/OFQ increases food intake in mammals, but its effect and mode of action in chicks are not fully known. We report herein that N/OFQ (5.0 nmol) has a stimulatory effect on food intake in layer-type chicks over a 2-h period after intracerebroventricular (icv) injection. Thirty minutes after central injection of N/OFQ (5.0 nmol) the concentration of agouti-related protein (AGRP) mRNA in the diencephalon increased, while cocaine- and amphetamine-regulated transcript (CART) mRNA decreased. However, concentrations of neuropeptide Y, proopiomelanocortin and glutamate decarboxylase mRNAs, and of catecholamines and excitatory amino acids were not affected. Simultaneous administration of alpha-melanocyte stimulating hormone (alpha-MSH: 1.0 pmol), a competitor of AGRP, completely blocked the orexigenic effect of N/OFQ (5.0 nmol). These data suggest that N/OFQ functions in layer chicks as an orexigenic peptide in the central nervous system, and that the AGRP and the CART neurons may mediate this function, as in mammals.


Asunto(s)
Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neuropéptidos/metabolismo , Péptidos Opioides/farmacología , Vasodilatadores/farmacología , Proteína Relacionada con Agouti/genética , Animales , Catecolaminas/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Pollos , Glutamato Descarboxilasa/genética , Hormonas/farmacología , Hipotálamo/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Neuropéptido Y/genética , Neuropéptidos/genética , Proopiomelanocortina/genética , alfa-MSH/farmacología , Nociceptina
15.
Neurogastroenterol Motil ; 20(8): 939-48, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18410266

RESUMEN

In this study, seeking further information on the role of the nociceptin/orphanin FQ (N/OFQ)-ergic system in normal and disturbed colonic motor function in rats, we compared the colonic effects of UFP-112, a novel highly potent agonist, with those of N/OFQ. When injected intracerebroventricularly (i.c.v.) and intraperitoneally (i.p.), UFP-112 and N/OFQ increased bead expulsion time in a statistically significant and dose-related manner and reduced the percentage of rats with castor oil-induced diarrhoea. UFP-112 showed greater efficacy, higher potency and longer-lasting inhibitory effects than N/OFQ, and pretreatment with UFP-101, a selective antagonist, blocked the N/OFQ analogue-induced responses in both tests. When injected i.c.v., UFP-112 and N/OFQ inhibited corticotrophin releasing factor- and restrain stress-stimulated faecal pellet excretion significantly and in a dose-related manner. Conversely, when injected peripherally both peptides significantly inhibited colonic propulsive motility but did so in a non-dose-related manner. In conclusion, these findings indicate that, in the rat, the central and peripheral N/OFQ systems have an inhibitory role in modulating distal colonic propulsive motility under physiological and pathological conditions. UFP-112 therefore promises to be a useful pharmacological tool for investigating the role of the N/OFQ system in motor functions in the distal colonic tract under physiological and pathological conditions.


Asunto(s)
Colon , Heces , Motilidad Gastrointestinal/efectos de los fármacos , Péptidos Opioides/farmacología , Animales , Aceite de Ricino/farmacología , Catárticos/farmacología , Colon/efectos de los fármacos , Colon/fisiología , Hormona Liberadora de Corticotropina/farmacología , Diarrea/inducido químicamente , Relación Dosis-Respuesta a Droga , Motilidad Gastrointestinal/fisiología , Tránsito Gastrointestinal/efectos de los fármacos , Tránsito Gastrointestinal/fisiología , Masculino , Ratas , Ratas Wistar , Restricción Física , Estrés Psicológico , Vasodilatadores/metabolismo , Nociceptina
16.
Endocrinology ; 148(10): 4993-5001, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17615146

RESUMEN

Orphanin FQ (OFQ), also known as nociceptin, is a member of the endogenous opioid peptide family that has been functionally implicated in the control of pain, anxiety, circadian rhythms, and neuroendocrine function. In the reproductive system, endogenous opioid peptides are involved in the steroid feedback control of GnRH pulses and the induction of the GnRH surge. The distribution of OFQ in the preoptic area and hypothalamus overlaps with GnRH, and in vitro evidence suggests that OFQ can inhibit GnRH secretion from hypothalamic fragments. Using the sheep as a model, we examined the potential anatomical colocalization between OFQ and GnRH using dual-label immunocytochemistry. Confocal microscopy revealed that approximately 93% of GnRH neurons, evenly distributed across brain regions, were also immunoreactive for OFQ. In addition, almost all GnRH fibers and terminals in the external zone of the median eminence, the site of neurosecretory release of GnRH, also colocalized OFQ. This high degree of colocalization suggested that OFQ might be functionally important in controlling reproductive endocrine events. We tested this possibility by examining the effects of intracerebroventricular administration of [Arg(14), Lys(15)] OFQ, an agonist to the OFQ receptor, on pulsatile LH secretion. The agonist inhibited LH pulse frequency in both luteal phase and ovariectomized ewes and suppressed pulse amplitude in the latter. The results provide in vivo evidence supporting a role for OFQ in the control of GnRH secretion and raise the possibility that it acts as part of an ultrashort, autocrine feedback loop controlling GnRH pulses.


Asunto(s)
Sistemas Neurosecretores/fisiología , Péptidos Opioides/fisiología , Reproducción/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Fase Luteínica/efectos de los fármacos , Hormona Luteinizante/metabolismo , Péptidos Opioides/agonistas , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Ovariectomía , Área Preóptica/metabolismo , Ovinos , Distribución Tisular , Nociceptina
17.
Brain Res ; 1078(1): 212-8, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16563360

RESUMEN

The neuropeptide nociceptin/orphanin FQ (N/OFQ), the endogenous agonist of the N/OFQ peptide receptor (NOP receptor), has been demonstrated to be involved in many physiological and pathological functions including pain regulation. In the present study, the involvement of N/OFQ-NOP receptor system in electroacupuncture (EA)-produced anti-hyperalgesia was investigated in rats with peripheral inflammation. Intrathecal (i.t.) administration of N/OFQ (15 nmol) or EA at acupoints GB30 and GB34 could significantly attenuate hyperalgesia which was induced by subcutaneously injecting complete Freund's adjuvant (CFA) into one hindpaw of rats, manifesting as decreased paw withdrawal latency (PWL) to the noxious thermal stimulus. The anti-nociceptive effect of N/OFQ or EA was significantly blocked by intrathecal injection of [Nphe(1)]nociceptin(1-13)NH(2) (20 nmol), a selective antagonist of the NOP receptor, indicating the NOP-receptor-mediated mechanism. Additionally, the combination of N/OFQ injection with EA treatment could enhance anti-hyperalgesia compared to that produced by each component alone. These findings suggested that the spinal N/OFQ-NOP system might be involved in EA analgesia, which may be one of the mechanisms underlying the anti-nociceptive effect of EA in rat's peripheral inflammatory pain.


Asunto(s)
Electroacupuntura/métodos , Hiperalgesia/etiología , Hiperalgesia/terapia , Inflamación/complicaciones , Péptidos Opioides/fisiología , Receptores Opioides/fisiología , Puntos de Acupuntura , Análisis de Varianza , Animales , Conducta Animal , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Adyuvante de Freund/toxicidad , Calor , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Péptidos Opioides/farmacología , Dimensión del Dolor/métodos , Fragmentos de Péptidos/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Tiempo de Reacción/efectos de la radiación , Receptor de Nociceptina , Nociceptina
18.
Am J Respir Crit Care Med ; 173(3): 271-5, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16239621

RESUMEN

RATIONALE: Nociceptin/orphanin FQ has been reported to inhibit capsaicin- and mechanically provoked cough in animal models, but the mechanism of this effect has not been elucidated. OBJECTIVES: The objectives of this study were to determine whether nociceptin inhibits acid-evoked cough in conscious animals and to evaluate the mechanism of this effect. METHODS: We tested the effect of nociceptin on acid-induced cough in conscious guinea pigs and acid-induced nerve activation in airway-specific vagal sensory neurons using calcium imaging techniques and the gramicidin-perforated patch clamp technique. MEASUREMENTS AND MAIN RESULTS: Nociceptin (3 mg/kg, intraperitoneal) effectively inhibited acid-evoked cough in guinea pigs by nearly 70%. Acid (pH 5) increased intracellular free calcium in acutely dissociated vagal jugular ganglionic neurons. The acid-induced increase in intracellular calcium was inhibited by a selective transient receptor potential vanilloid-1 antagonist, 5-iodo-resiniferatoxin (1 microM, approximately 80% reduction). The inhibitory effect of 5-iodo-resiniferatoxin on acid-induced increases in calcium was mimicked by nociceptin (0.1 microM). In gramicidin-perforated patch clamp recordings on airway-specific capsaicin-sensitive jugular ganglion neurons, acid (pH 5) induced two distinct inward currents. A transient current was evoked that was inhibited by amiloride and a sustained current was evoked that was inhibited by 5-iodo-resiniferatoxin. Nociceptin selectively inhibited only the sustained component of acid-induced inward current. CONCLUSION: These results indicate that the inhibitory effect of nociceptin on acid-induced cough may result from a direct inhibitory effect on peripheral C-fiber activity caused by the selective inhibition of acid-induced transient receptor potential vanilloid-1 activation.


Asunto(s)
Tos/prevención & control , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Receptores Opioides/agonistas , Sistema Respiratorio/efectos de los fármacos , Animales , Cuerpos Aórticos/efectos de los fármacos , Capsaicina , Ácido Cítrico , Tos/inducido químicamente , Glomo Yugular/efectos de los fármacos , Cobayas , Masculino , Neuronas Aferentes/efectos de los fármacos , Sistema Respiratorio/inervación , Receptor de Nociceptina , Nociceptina
19.
Acta Pharmacol Sin ; 26(9): 1039-44, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16115368

RESUMEN

AIM: To investigate effect of the nociceptin/orphanin FQ (OFQ) on hypothalamus gonadotropin-releasing hormone (GnRH) release in ovariectomized (OVX) rats. METHODS: GnRH radioimmunoassay (RIA) was used to study the effect of OFQ on GnRH release in hypothalamus slices in vitro. Push-pull perfusion and intracerebroventicular (icv) injection were used to examine the effect of OFQ on GnRH release in the hypothalamus medial preoptic area (POA) in vivo. Ovariectomies were performed on female Sprague-Dawley rats, and their plasma luteinizing hormone (LH) levels were measured after icv injection of OFQ with or without [Nphe1]NC(1-13)NH2, a competitive antagonist of opioid receptor-like1 receptor (ORL1 receptor). Reverse transcription-polymerase chain reaction (RT-PCR) was used to investigate the expression of the ORL1 receptor in rat pituitary. RESULTS: GnRH release from hypothalamus slices was inhibited 90 min after the administration of 2 mmol/L and 20 mmol/L OFQ (P<0.05). Accordingly, GnRH release from hypothalamus POA was also significantly reduced by the injection of 0.2 mmol/L and 2 mmol/L OFQ. Plasma LH levels were also decreased significantly 2 h after icv injection of 20 nmol OFQ in OVX rats (P<0.05) and this effect could be abolished by pretreatment with 20 nmol [Nphe1]NC(1-13)NH2, that is, NC13. More interestingly, plasma LH levels in OVX rats increased markedly 2 h after icv injection of 100 nmol and 200 nmol NC13. RT-PCR analysis further revealed that the ORL1 receptor was not expressed in the pituitary of OVX rats. CONCLUSION: Central administration of nociceptin/orphanin FQ might inhibit the release of hypothalamic GnRH and decrease the plasma LH levels through ORL1 receptors in OVX rats.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Péptidos Opioides/farmacología , Hipófisis/metabolismo , Receptores Opioides/metabolismo , Animales , Femenino , Hormona Luteinizante/sangre , Péptidos Opioides/administración & dosificación , Ovariectomía , Área Preóptica/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Nociceptina , Nociceptina
20.
Endocrinology ; 146(8): 3526-34, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15890775

RESUMEN

Nociceptin or orphanin FQ (N/OFQ) and its receptor NOP1 are expressed in hypothalamic nuclei involved in energy homeostasis. N/OFQ administered by intracerebroventricular or arcuate nucleus (ARC) injection increases food intake in satiated rats. The mechanisms by which N/OFQ increases food intake are unknown. We hypothesized that N/OFQ may regulate hypothalamic neurons containing peptides involved in the control of food intake such as cocaine- and amphetamine-regulated transcript (CART), alphaMSH, neuropeptide Y (NPY), and agouti-related protein (AgRP). We investigated the ability of N/OFQ to alter the release of CART, alphaMSH, NPY, and AgRP using ex vivo medial basal hypothalamic explants. Incubation of hypothalamic explants with N/OFQ (1, 10, 100 nM) resulted in significant changes in CART and AgRP release. One hundred nanomoles N/OFQ caused a 33% decrease in release of CART (55-102) immunoreactivity (IR) and increased release of AgRP-IR to 163% but produced no change in either alphaMSH-IR or NPY-IR. Double immunocytochemistry/in situ hybridization demonstrated that CART-IR and NOP1 mRNA are colocalized throughout the hypothalamus, in particular in the paraventricular nucleus, lateral hypothalamus, zona incerta, and ARC, providing an anatomical basis for N/OFQ action on CART release. Dual in situ hybridization demonstrated that AgRP neurons in the ARC also express the NOP1 receptor. Our data suggest that nociceptin via the NOP1 receptor may increase food intake by decreasing the release of the anorectic peptide CART and increasing the release of the orexigenic peptide AgRP.


Asunto(s)
Regulación de la Expresión Génica , Hipotálamo/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Péptidos Opioides/genética , Péptidos Opioides/farmacología , Hormonas Peptídicas/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/fisiología , Ingestión de Energía , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intercelular , Masculino , Neuropéptido Y/metabolismo , Técnicas de Cultivo de Órganos , Hormonas Peptídicas/metabolismo , Ratas , Ratas Wistar , Receptores Opioides , Ribonucleasas , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA