Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Behav Brain Res ; 372: 112043, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31226311

RESUMO

Kissorphin (KSO) is a new peptide derived from kisspeptin-10. Previous study has indicated that this peptide displays neuropeptide FF (NPFF)-like anti-opioid activity. Herein, we examined the influence of KSO (1; 3, and 10 nmol, intravenously [i.v.]), on the rewarding action of morphine (5 mg/kg, intraperitoneally [i.p.]), using the unbiased design of the conditioned place preference (CPP) paradigm in rats. To test the effect of KSO on the acquisition of morphine-induced CPP, KSO and morphine were co-injected during conditioning with no drugs treatment on the test day. To investigate the effect of KSO on the expression of morphine-induced CPP, morphine alone was given during the conditioning phase (1 × 3 days) and KSO was administered 5 min prior to the placement in the CPP apparatus on the test day. To estimate the influence of KSO on the reinstatement of morphine-induced CPP, KSO was given 5 min before a priming dose of morphine (5 mg/kg, i.p.) on the reinstatement test day. The results show that KSO inhibited the acquisition, expression and reinstatement of morphine-induced CPP. The strongest effect of KSO was observed at the dose of 10 nmol (acquisition and reinstatement) or 1 nmol (expression). KSO given alone, neither induced place preference, nor aversion. Furthermore, the morphine-modulating effects of KSO were markedly antagonized by pretreatment with RF9 (10 nmol, i.v.), the NPFF receptors selective antagonist. Thus, KSO inhibited the morphine-induced CPP mainly by involving specific activation of NPFF receptors. Overall, these data further support the anti-opioid character of KSO.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Kisspeptinas/farmacologia , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Kisspeptinas/metabolismo , Masculino , Morfina/farmacologia , Ratos , Ratos Wistar , Receptores de Neuropeptídeos/metabolismo , Receptores de Neuropeptídeos/fisiologia , Recompensa
2.
Pharmacol Ther ; 196: 59-78, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30439454

RESUMO

Obesity is a chronic multifactorial disease, characterized by an excessive accumulation of adipose tissue. It is usually the result of excessive food intake and/or low energy expenditure. Obesity can be triggered by lifestyle, nutritional, genetic, environmental, hormonal and psychological factors. Several strategies are used to treat obesity, including dietary reeducation, with balanced food intake, increased physical exercise, in order to promote energy expenditure and to overcome the insufficiency in weight reduction by other strategies, and administration of drugs. However, these medications are associated to undesirable side effects, resulting in a high withdrawal rate. Several studies have been focused on the development of compounds that act in the hypothalamic region where the center of the regulation of hunger and satiety is located. Some of them target the activity of endogenous peptides, such as ghrelin pancreatic polypeptide, peptide YY and neuropeptide Y, as well as their receptors. This review addresses the importance of understanding the neuropeptide/peptide hormones and their receptors for the development of novel anti-obesity compounds that may aid in weight reduction as a promising alternative for the treatment of obesity.


Assuntos
Obesidade/fisiopatologia , Receptores de Neuropeptídeos/fisiologia , Animais , Humanos , Fome , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Resposta de Saciedade
3.
Cephalalgia ; 39(13): 1661-1674, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-29989427

RESUMO

INTRODUCTION: The trigeminal ganglion is unique among the somatosensory ganglia regarding its topography, structure, composition and possibly some functional properties of its cellular components. Being mainly responsible for the sensory innervation of the anterior regions of the head, it is a major target for headache research. One intriguing question is if the trigeminal ganglion is merely a transition site for sensory information from the periphery to the central nervous system, or if intracellular modulatory mechanisms and intercellular signaling are capable of controlling sensory information relevant for the pathophysiology of headaches. METHODS: An online search based on PubMed was made using the keyword "trigeminal ganglion" in combination with "anatomy", "headache", "migraine", "neuropeptides", "receptors" and "signaling". From the relevant literature, further references were selected in view of their relevance for headache mechanisms. The essential information was organized based on location and cell types of the trigeminal ganglion, neuropeptides, receptors for signaling molecules, signaling mechanisms, and their possible relevance for headache generation. RESULTS: The trigeminal ganglion consists of clusters of sensory neurons and their peripheral and central axon processes, which are arranged according to the three trigeminal partitions V1-V3. The neurons are surrounded by satellite glial cells, the axons by Schwann cells. In addition, macrophage-like cells can be found in the trigeminal ganglion. Neurons express various neuropeptides, among which calcitonin gene-related peptide is the most prominent in terms of its prevalence and its role in primary headaches. The classical calcitonin gene-related peptide receptors are expressed in non-calcitonin gene-related peptide neurons and satellite glial cells, although the possibility of a second calcitonin gene-related peptide receptor in calcitonin gene-related peptide neurons remains to be investigated. A variety of other signal molecules like adenosine triphosphate, nitric oxide, cytokines, and neurotrophic factors are released from trigeminal ganglion cells and may act at receptors on adjacent neurons or satellite glial cells. CONCLUSIONS: The trigeminal ganglion may act as an integrative organ. The morphological and functional arrangement of trigeminal ganglion cells suggests that intercellular and possibly also autocrine signaling mechanisms interact with intracellular mechanisms, including gene expression, to modulate sensory information. Receptors and neurotrophic factors delivered to the periphery or the trigeminal brainstem can contribute to peripheral and central sensitization, as in the case of primary headaches. The trigeminal ganglion as a target of drug action outside the blood-brain barrier should therefore be taken into account.


Assuntos
Cefaleia/fisiopatologia , Gânglio Trigeminal/fisiopatologia , Trifosfato de Adenosina/metabolismo , Vias Aferentes/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina/uso terapêutico , Citocinas/metabolismo , Cefaleia/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Transtornos de Enxaqueca/tratamento farmacológico , Transtornos de Enxaqueca/patologia , Transtornos de Enxaqueca/fisiopatologia , Fatores de Crescimento Neural/metabolismo , Neuropeptídeos/fisiologia , Óxido Nítrico/metabolismo , Nociceptividade/fisiologia , Ratos , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/fisiologia , Receptores de Neuropeptídeos/fisiologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/patologia
4.
Neurosci Lett ; 684: 98-103, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-29981879

RESUMO

Considerable evidence suggests the Neuropeptide FF (NPFF) and related peptides exert pro-nociceptive and anti-opiate actions, particularly at the supra-spinal level, which may contribute to opiate dependence. The FF1 receptor subtype appears to be primarily responsible for anti-opiate effects. In contrast, stimulation of the FF2 receptor primarily induces pro-opiate effects. AC-262620 is a small molecule, systemically active, selective FF1 receptor antagonist. An initial experiment showed that 10 mg/kg i.p. AC-262620 significantly reduced subsequent naloxone-precipitated somatically expressed withdrawal signs in rats infused s.c. for seven days with 0.3 mg/kg/hr morphine sulfate. A second experiment showed that the same dose of AC-262620 significantly reduced subsequent spontaneous withdrawal signs 23.75 h after termination of seven days s.c. infusion of 0.6 mg/kg/hr morphine sulfate. Chronic nicotine intake may contribute to dependence by overstimulating opiate receptors through release of opiate peptides. By analogy to opiate dependence, it was hypothesized that FF1 receptor activation contributes to nicotine dependence and withdrawal syndrome. AC-262620 significantly reduced somatically expressed withdrawal signs precipitated by the nicotinic antagonist mecamylamine in rats infused for seven days with nicotine bitartrate. Taken together, these findings suggest that NPFF or related neuropeptides contribute to opiate, as well as nicotine, dependence and withdrawal syndrome through the FF1 receptor.


Assuntos
Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Receptores de Neuropeptídeos/antagonistas & inibidores , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Tabagismo/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Animais , Masculino , Morfina/efeitos adversos , Nicotina/efeitos adversos , Transtornos Relacionados ao Uso de Opioides/psicologia , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/fisiologia , Síndrome de Abstinência a Substâncias/psicologia , Tabagismo/psicologia
5.
ACS Chem Biol ; 13(5): 1343-1352, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29543428

RESUMO

Neuropeptides in several animals undergo an unusual post-translational modification, the isomerization of an amino acid residue from the l-stereoisomer to the d-stereoisomer. The resulting d-amino acid-containing peptide (DAACP) often displays biological activity higher than that of its all-l-residue analogue, with the d-residue being critical for function in many cases. However, little is known about the full physiological roles played by DAACPs, and few studies have examined the interaction of DAACPs with their cognate receptors. Here, we characterized the signaling of several DAACPs derived from a single neuropeptide prohormone, the Aplysia californica achatin-like neuropeptide precursor (apALNP), at their putative receptor, the achatin-like neuropeptide receptor (apALNR). We first used quantitative polymerase chain reaction and in situ hybridization experiments to demonstrate receptor ( apALNR) expression throughout the central nervous system; on the basis of the expression pattern, we identified novel physiological functions that may be mediated by apALNR. To gain insight into ligand signaling through apALNR, we created a library of native and non-native neuropeptide analogues derived from apALNP (the neuropeptide prohormone) and evaluated them for activity in cells co-transfected with apALNR and the promiscuous Gα subunit Gα-16. Several of these neuropeptide analogues were also evaluated for their ability to induce circuit activity in a well-defined neural network associated with feeding behavior in intact ganglia from Aplysia. Our results reveal the specificity of apALNR and provide strong evidence that this receptor mediates diverse physiological functions throughout the central nervous system. Finally, we show that some native apALNP-derived DAACPs exhibit enhanced stability toward endogenous proteases, suggesting that the d-residues in these DAACPs may increase the peptide lifetime, in addition to influencing receptor specificity, in the nervous system. Ultimately, these studies provide insight into signaling at one of the few known DAACP-specific receptors and advance our understanding of the roles that l- to d-residue isomerization play in neuropeptide signaling.


Assuntos
Aminoácidos/análise , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Sequência de Aminoácidos , Animais , Aplysia , Sistema Nervoso Central/metabolismo , Ligantes , Peptídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Neuropeptídeos/metabolismo
6.
Gen Comp Endocrinol ; 261: 97-103, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29481768

RESUMO

Neuropeptide B (NPB) is an endogenous ligand for the orphan G protein-coupled receptors NPBWR1 (GPR7) and NPBWR2 (GPR8). Some reports have investigated the role of NPB in the regulation of feeding, energy metabolism and hormone secretion in many species. However, few papers reported the physiological function of NPB in the pig. In this study, we cloned and sequenced the NPB mRNA from a pig, which was found to consist of 123 bases. NPB mRNA expression was detected in central and peripheral tissues by the quantitative fluorescence method. The results showed that NPB mRNA expression was higher in hippocampus, cerebellum, spinal cord, thymus, tonsil, duodenum, cecum, colon, ovary and testis. The distribution of NPB suggested that it may be involved in the regulation of reproductive functions in the pig. Subsequently, the expression and distribution of NPBWR1 and NPBWR2 were found in Leydig cells and ovarian granular cells. We then investigated the direct effect of NPB on pig reproductive cells in vitro. The results showed that different concentrations of NPB (10-12, 10-10, 10-8 and 10-6 M) promoted the secretion of testosterone in Leydig cells in concentration-dependent manner. Different doses of NPB could promote the secretion of progesterone in ovarian granulosa cells in dose-dependent manner. Low concentrations of NPB (10-8 and 10-10 M) promoted estradiol secretion, but high concentrations of NPB (10-6 M) inhibited its secretion. All the results suggested that the NPB/NPBWR1 or NPBWR2 system may play a role in modulating the reproductive activity in the pig.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Gônadas/efeitos dos fármacos , Gônadas/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Suínos/genética , Animais , Sequência de Bases , Clonagem Molecular , Feminino , Gônadas/citologia , Masculino , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/fisiologia , Reprodução/genética , Via Secretória/efeitos dos fármacos , Suínos/metabolismo
7.
J Neurosci ; 37(50): 12214-12225, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29118105

RESUMO

Neuropeptides, such as neuropeptide S (NPS) and oxytocin (OXT), represent potential options for the treatment of anxiety disorders due to their potent anxiolytic profile. In this study, we aimed to reveal the mechanisms underlying the behavioral action of NPS, and present a chain of evidence that the effects of NPS within the hypothalamic paraventricular nucleus (PVN) are mediated via actions on local OXT neurons in male Wistar rats. First, retrograde studies identified NPS fibers originating in the brainstem locus coeruleus, and projecting to the PVN. FACS identified prominent NPS receptor expression in PVN-OXT neurons. Using genetically encoded calcium indicators, we further demonstrated that NPS reliably induces a transient increase in intracellular Ca2+ concentration in a subpopulation of OXT neurons, an effect mediated by NPS receptor. In addition, intracerebroventricular (i.c.v.) NPS evoked a significant somatodendritic release of OXT within the PVN as assessed by microdialysis in combination with a highly sensitive radioimmunoassay. Finally, we could show that the anxiolytic effect of NPS seen after i.c.v. or intra-PVN infusion requires responsive OXT neurons of the PVN and locally released OXT. Thus, pharmacological blockade of OXT receptors as well as chemogenetic silencing of OXT neurons within the PVN prevented the effect of synthetic NPS. In conclusion, our results indicate a significant role of the OXT system in mediating the effects of NPS on anxiety, and fill an important gap in our understanding of brain neuropeptide interactions in the context of regulation of emotional behavior within the hypothalamus.SIGNIFICANCE STATEMENT Given the rising scientific interest in neuropeptide research in the context of emotional and stress-related behaviors, our findings demonstrate a novel intrahypothalamic mechanism involving paraventricular oxytocin neurons that express the neuropeptide S receptor. These neurons respond with transient Ca2+ increase and somatodendritic oxytocin release following neuropeptide S stimulation. Thereby, oxytocin neurons seem essential for neuropeptide S-induced anxiolysis, as this effect was blocked by pharmacological and chemogenetic inhibition of the oxytocin system.


Assuntos
Ansiedade/fisiopatologia , Neuropeptídeos/fisiologia , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptores de Ocitocina/fisiologia , Animais , Transporte Axonal , Proteínas de Bactérias/análise , Sinalização do Cálcio/fisiologia , Dependovirus/genética , Comportamento Exploratório/efeitos dos fármacos , Genes Reporter , Vetores Genéticos , Proteínas Luminescentes/análise , Masculino , Microdiálise , Atividade Motora/efeitos dos fármacos , Neuropeptídeos/farmacologia , Ocitocina/agonistas , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Neuropeptídeos/efeitos dos fármacos , Receptores de Neuropeptídeos/fisiologia , Receptores de Ocitocina/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Proteína Vermelha Fluorescente
8.
Vet Immunol Immunopathol ; 188: 34-47, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28615126

RESUMO

Vasoactive peptides are key early mediators of inflammation released through activation of different enzymatic systems. The mammalian kinin-kallikrein (K-KLK) system produces bradykinin (BK) through proteolytic cleavage of a kininogen precursor by enzymes named kallikreins. BK acts through specific ubiquitous G-protein coupled receptors (B1R and B2R) to participate in physiological processes and inflammatory responses, such as activation of mononuclear phagocytes. In chickens, the BK-like nonapeptide ornithokinin (OK) has been shown to promote intracellular calcium increase in embryonic fibroblasts and to be vasodilatory in vivo. Also, one of its receptors (B2R) was already cloned. However, the participation of chicken K-KLK system components in the inflammatory response remains unknown and was therefore investigated. We first showed that B1R, B2R and kininogen 1 (KNG1) are expressed in unstimulated chicken tissues and macrophages. We next showed that chicken B1R and B2R are expressed at transcript and protein levels in chicken macrophages and are upregulated by E. coli LPS or avian pathogenic E. coli (APEC) infection. Interestingly, exogenous OK induced internalization and degradation of OK receptors protein, notably B2R. Also, OK induced intracellular calcium increase and potentiated zymosan-induced ROS production and Dextran-FITC endocytosis by chicken macrophages. Exogenous OK itself did not promote APEC killing and had no pro-inflammatory effect. However, when combined with LPS or APEC, OK upregulated cytokine/chemokine gene expression and NO production by chicken macrophages. This effect was not blocked by canonical non-peptide B1R or B2R receptor antagonists but was GPCR- and PI3K/Akt-dependent. In vivo, pulmonary colibacillosis led to upregulation of OK receptors expression in chicken lungs and liver. Also, colibacillosis led to significant upregulation of OK precursor KNG1 expression in liver and in cultured hepatocytes (LMH). We therefore provide hitherto unknown information on how OK and its receptors are involved in inflammation and infection in chickens.


Assuntos
Bradicinina/análogos & derivados , Inflamação/veterinária , Cininas/fisiologia , Doenças das Aves Domésticas/imunologia , Receptores de Neuropeptídeos/fisiologia , Animais , Bradicinina/fisiologia , Galinhas/imunologia , Escherichia coli/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Doenças das Aves Domésticas/metabolismo
9.
Neuropharmacology ; 108: 364-72, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26970017

RESUMO

Mounting evidences indicate the functional interactions between neuropeptide FF (NPFF) and opioids, including the endogenous opioids. In the present work, EN-9, a chimeric peptide containing the functional domains of the endogenous opioid endomorphin-2 (EM-2) and NPFF, was synthesized and pharmacologically characterized. In vitro cAMP assay demonstrated that EN-9 was a multifunctional agonist of κ-opioid, NPFF1 and NPFF2 receptors. In the mouse tail-flick test, intracerebroventricularly (i.c.v.) administration of EN-9 produced significant antinociception with an ED50 value of 13.44 nmol, which lasted longer than that of EM-2. In addition, EN-9 induced potent antinociception after both intravenous (i.v.) and subcutaneous (s.c.) injection. Furthermore, the experiments using the antagonists of opioid and NPFF receptors indicated that the central antinociception of EN-9 was mainly mediated by κ-opioid receptor, independently on NPFF receptors. Notably, the central antinociception of EN-9 was not reduced over a period of 6 days repeated i.c.v. injection. Repeated i.c.v. administration of EN-9 with the NPFF1 and NPFF2 receptors antagonist RF9 resulted in a progressive loss of analgesic potency, consistent with the development of tolerance. Moreover, central administration of EN-9 induced the place conditioning aversion only at a high dose of 60 nmol, but not at low doses. At supraspinal level, only high dose of EN-9 (60 nmol, i.c.v.) inhibited gastrointestinal transit via NPFF receptors. Similarly, systemic administration of EN-9 also inhibited gastrointestinal transit at high doses (10 and 30 mg/kg, i.v.). Taken together, the multifunctional agonist of κ-opioid and NPFF receptors EN-9 produced a potent, non-tolerance forming antinociception with limited side effects.


Assuntos
Analgésicos Opioides/administração & dosagem , Oligopeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Analgésicos/administração & dosagem , Analgésicos/química , Analgésicos Opioides/química , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/fisiologia , Células HEK293 , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Oligopeptídeos/química , Medição da Dor/métodos , Fragmentos de Peptídeos/química , Peptídeos/química , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/fisiologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/fisiologia
10.
Brain Struct Funct ; 221(6): 3327-36, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26323488

RESUMO

Our previous studies have demonstrated that neuropeptide S (NPS), via selective activation of the neurons bearing NPS receptor (NPSR) in the olfactory cortex, facilitates olfactory function. High level expression of NPSR mRNA in the subiculum complex of hippocampal formation suggests that NPS-NPSR system might be involved in the regulation of olfactory spatial memory. The present study was undertaken to investigate effects of NPS on the scopolamine- or MK801-induced impairment of olfactory spatial memory using computer-assisted 4-hole-board spatial memory test, and by monitoring Fos expression in the subiculum complex in mice. In addition, dual-immunofluorescence microscopy was employed to identify NPS-induced Fos-immunereactive (-ir) neurons that also bear NPSR. Intracerebroventricular administration of NPS (0.5 nmol) significantly increased the number of visits to switched odorants in recall trial in mice suffering from odor-discriminating inability induced by scopolamine, a selective muscarinic cholinergic receptor antagonist, or MK801, a N-methyl-D-aspartate receptor antagonist, after training trials. The improvement of olfactory spatial memory by NPS was abolished by the NPSR antagonist [D-Val(5)]NPS (40 nmol). Ex vivo c-Fos and NPSR immunohistochemistry revealed that, as compared with vehicle-treated mice, NPS markedly enhanced Fos expression in the subiculum complex encompassing the subiculum (S), presubiculum (PrS) and parasubiculum (PaS). The percentages of Fos-ir neurons that also express NPSR were 91.3, 86.5 and 90.0 % in the S, PrS and PaS, respectively. The present findings demonstrate that NPS, via selective activation of the neurons bearing NPSR in the subiculum complex, ameliorates olfactory spatial memory impairment induced by scopolamine and MK801 in mice.


Assuntos
Hipocampo/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Percepção Olfatória/fisiologia , Receptores de Neuropeptídeos/fisiologia , Memória Espacial/fisiologia , Animais , Maleato de Dizocilpina/administração & dosagem , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neuropeptídeos/administração & dosagem , Odorantes , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Neuropeptídeos/metabolismo , Escopolamina/administração & dosagem , Memória Espacial/efeitos dos fármacos
11.
Endocrinology ; 155(8): 2953-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24823392

RESUMO

RF-amide-related peptide-3 (RFRP-3), the mammalian ortholog of the avian gonadotropin-inhibiting hormone (GnIH), operates via the NPFF1 receptor (NPFF1R) to repress the reproductive axis, therefore acting as counterpart of the excitatory RF-amide peptide, kisspeptin (ligand of Gpr54). In addition, RFRP-3 modulates feeding and might contribute to the integrative control of energy homeostasis and reproduction. Yet, the experimental evidence supporting these putative functions is mostly indirect, and the physiological roles of RFRP-3 remain debatable and obscured by the lack of proper analytical tools and models. To circumvent these limitations, we characterize herein the first mouse line with constitutive inactivation of NPFF1R. Ablation of NPFF1R did not compromise fertility; rather, litters from NPFF1R null mice were larger than those from wild-type animals. Pubertal timing was not altered in NPFF1R deficient mice; yet, pre-pubertal knockout (KO) males displayed elevated LH levels, which normalized after puberty. Adult NPFF1R null male mice showed increased Kiss1 expression in the hypothalamic arcuate nucleus, higher serum FSH levels, and enhanced LH responses to GnRH. However, genetic elimination of NPFF1R was unable to reverse the state of hypogonadism caused by the lack of kisspeptin signaling, as revealed by double NPFF1R/Gpr54 KO mice. NPFF1R null mice displayed altered feedback responses to gonadal hormone withdrawal. In addition, metabolic challenges causing gonadotropin suppression, such as short-term fasting and high-fat diet, were less effective in dampening LH secretion in NPFF1R-deficient male mice, suggesting that absence of this inhibitory pathway partially prevented gonadotropin suppression by metabolic stress. Our data are the first to document the impact of elimination of GnIH signaling on reproductive parameters and their modulation by metabolic challenges. Whereas, in keeping with its inhibitory role, the NPFF1R pathway seems dispensable for preserved puberty and fertility, our results surface different alterations due to the lack of GnIH signaling that prominently include changes in the sensitivity to fasting- and obesity-associated hypogonadotropism.


Assuntos
Gonadotropinas/fisiologia , Tamanho da Ninhada de Vivíparos , Neuropeptídeos/fisiologia , Receptores de Neuropeptídeos/fisiologia , Maturidade Sexual , Animais , Jejum , Retroalimentação Fisiológica , Feminino , Fertilidade , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Fenótipo , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Maturidade Sexual/genética , Estresse Fisiológico/genética
12.
J Immunol ; 191(1): 345-52, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23698749

RESUMO

Human ß-defensins (hBDs) stimulate degranulation in rat peritoneal mast cells in vitro and cause increased vascular permeability in rats in vivo. In this study, we sought to determine whether hBDs activate murine and human mast cells and to delineate the mechanisms of their regulation. hBD2 and hBD3 did not induce degranulation in murine peritoneal or bone marrow-derived mast cells (BMMC) in vitro and had no effect on vascular permeability in vivo. By contrast, these peptides induced sustained Ca(2+) mobilization and substantial degranulation in human mast cells, with hBD3 being more potent. Pertussis toxin (PTx) had no effect on hBD-induced Ca(2+) mobilization, but La(3+) and 2-aminoethoxydiphenyl borate (a dual inhibitor of inositol 1,4,5-triphosphate receptor and transient receptor potential channels) caused substantial inhibition of this response. Interestingly, degranulation induced by hBDs was substantially inhibited by PTx, La(3+), or 2-aminoethoxydiphenyl borate. Whereas human mast cells endogenously express G protein-coupled receptor, Mas-related gene X2 (MrgX2), rat basophilic leukemia, RBL-2H3 cells, and murine BMMCs do not. Silencing the expression of MrgX2 in human mast cells inhibited hBD-induced degranulation, but had no effect on anaphylatoxin C3a-induced response. Furthermore, ectopic expression of MrgX2 in RBL-2H3 and murine BMMCs rendered these cells responsive to hBDs for degranulation. This study demonstrates that hBDs activate human mast cells via MrgX2, which couples to both PTx-sensitive and insensitive signaling pathways most likely involving Gαq and Gαi to induce degranulation. Furthermore, murine mast cells are resistant to hBDs for degranulation, and this reflects the absence of MrgX2 in these cells.


Assuntos
Mastócitos/imunologia , Mastócitos/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , beta-Defensinas/fisiologia , Animais , Sinalização do Cálcio/genética , Sinalização do Cálcio/imunologia , Degranulação Celular/genética , Degranulação Celular/imunologia , Linhagem Celular , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Ratos , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/biossíntese , Receptores de Neuropeptídeos/genética
13.
Neuropsychopharmacology ; 38(7): 1352-64, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23358240

RESUMO

Neuropeptide Y (NPY) administration into the basolateral amygdala (BLA) decreases anxiety-like behavior, mediated in part through the Y1 receptor (Y1R) isoform. Activation of Y1Rs results in G-protein-mediated reduction of cAMP levels, which results in reduced excitability of amygdala projection neurons. Understanding the mechanisms linking decreased cAMP levels to reduced excitability in amygdala neurons is important for identifying novel anxiolytic targets. We studied the intracellular mechanisms of activation of Y1Rs on synaptic transmission in the BLA. Activating Y1Rs by [Leu(31),Pro(34)]-NPY (L-P NPY) reduced the amplitude of evoked NMDA-mediated excitatory postsynaptic currents (eEPSCs), without affecting AMPA-mediated eEPSCs, but conversely increased the amplitude of GABAA-mediated evoked inhibitory postsynaptic currents (eIPSCs). Both effects were abolished by the Y1R antagonist, PD160170. Intracellular GDP-ß-S, or pre-treatment with either forskolin or 8Br-cAMP, eliminated the effects of L-P NPY on both NMDA- and GABAA-mediated currents. Thus, both the NMDA and GABAA effects of Y1R activation in the BLA are G-protein-mediated and cAMP-dependent. Pipette inclusion of protein kinase A (PKA) catalytic subunit blocked the effect of L-P NPY on GABAA-mediated eIPSCs, but not on NMDA-mediated eEPSCs. Conversely, activating the exchange protein activated by cAMP (Epac) with 8CPT-2Me-cAMP blocked the effect of L-P NPY on NMDA-mediated eEPSCs, but not on GABAA-mediated eIPSCs. Thus, NPY regulates amygdala excitability via two signal-transduction events, with reduced PKA activity enhancing GABAA-mediated eIPSCs and Epac deactivation reducing NMDA-mediated eEPSCs. This multipathway regulation of NMDA- and GABAA-mediated currents may be important for NPY plasticity and stress resilience in the amygdala.


Assuntos
Tonsila do Cerebelo/fisiologia , Neuropeptídeo Y/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Transdução de Sinais/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Aminoquinolinas/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Ansiolíticos/farmacologia , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Fatores de Troca do Nucleotídeo Guanina/efeitos dos fármacos , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neuropeptídeo Y/administração & dosagem , Neuropeptídeo Y/análogos & derivados , Neuropeptídeo Y/antagonistas & inibidores , Neuropeptídeo Y/farmacologia , Ratos , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/fisiologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/fisiologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sulfonas/farmacologia , Tionucleotídeos/farmacologia
14.
J Neurosci ; 32(25): 8509-20, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22723691

RESUMO

Establishing the pattern of expression of transmitters and peptides as well as their receptors in different neuronal types is crucial for understanding the circuitry in various regions of the brain. Previous studies have demonstrated that the transmitter and peptide phenotypes in mouse dorsal spinal cord neurons are determined by the transcription factors Tlx1/3 and Ptf1a. Here we show that these transcription factors also determine the expression of two distinct sets of transmitter and peptide receptor genes in this region. We have screened the expression of 78 receptor genes in the spinal dorsal horn by in situ hybridization. We found that receptor genes Gabra1, Gabra5, Gabrb2, Gria3, Grin3a, Grin3b, Galr1, and Npy1r were preferentially expressed in Tlx3-expressing glutamatergic neurons and their derivatives, and deletion of Tlx1 and Tlx3 resulted in the loss of expression of these receptor genes. Furthermore, we obtained genetic evidence that Tlx3 uses distinct pathways to control the expression of receptor genes. We also found that receptor genes Grm3, Grm4, Grm5, Grik1, Grik2, Grik3, and Sstr2 were mainly expressed in Pax2-expressing GABAergic neurons in the spinal dorsal horn, and their expression in this region was abolished or markedly reduced in Ptf1a and Pax2 deletion mutant mice. Together, our studies indicate that Tlx1/3 and Ptf1a, the key transcription factors for fate determination of glutamatergic and GABAergic neurons in the dorsal spinal cord, are also responsible for controlling the expression of two distinct sets of transmitter and peptide receptor genes.


Assuntos
Proteínas de Homeodomínio/fisiologia , Receptores de Neuropeptídeos/fisiologia , Receptores de Neurotransmissores/fisiologia , Medula Espinal/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Animais Geneticamente Modificados , Contagem de Células , Proteínas de Homeodomínio/genética , Hibridização In Situ , Camundongos , Camundongos Knockout , Neurotensina/metabolismo , Fator de Transcrição PAX2/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Reação em Cadeia da Polimerase , Receptores da Colecistocinina/genética , Receptores de Glutamato/genética , Receptores de Neuropeptídeos/genética , Receptores de Neurotransmissores/genética , Medula Espinal/metabolismo , Fatores de Transcrição/genética , Proteína Vesicular 1 de Transporte de Glutamato/genética , Ácido gama-Aminobutírico/fisiologia
15.
Physiol Behav ; 107(5): 733-42, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22554617

RESUMO

BACKGROUND: Although the hypothalamic orexin system is known to regulate appetitive behaviors and promote wakefulness and arousal (Sakurai, 2007 [56]), this system may also be important in adaptive and pathological anxiety/stress responses (Suzuki et al., 2005 [4]). In a recent study, we demonstrated that CSF orexin levels were significantly higher in patients experiencing panic attacks compared to non-panicking depressed subjects (Johnson et al., 2010 [9]). Furthermore, genetically silencing orexin synthesis or blocking orexin 1 receptors attenuated lactate-induced panic in an animal model of panic disorder. Therefore, in the present study, we tested if orexin (ORX) modulates panic responses and brain pathways activated by two different panicogenic drugs. METHODS: We conducted a series of pharmacological, behavioral, physiological and immunohistochemical experiments to study the modulation by the orexinergic inputs of anxiety behaviors, autonomic responses, and activation of brain pathways elicited by systemic injections of anxiogenic/panicogenic drugs in rats. RESULTS: We show that systemic injections of two different anxiogenic/panicogenic drugs (FG-7142, an inverse agonist at the benzodiazepine site of the GABA(A) receptor, and caffeine, a nonselective competitive adenosine receptor antagonist) increased c-Fos induction in a specific subset of orexin neurons located in the dorsomedial/perifornical (DMH/PeF) but not the lateral hypothalamus. Pretreating rats with an orexin 1 receptor antagonist attenuated the FG-7142-induced anxiety-like behaviors, increased heart rate, and neuronal activation in key panic pathways, including subregions of the central nucleus of the amygdala, bed nucleus of the stria terminalis, periaqueductal gray and in the rostroventrolateral medulla. CONCLUSION: Overall, the data here suggest that the ORX neurons in the DMH/PeF region are critical to eliciting coordinated panic responses and that ORX1 receptor antagonists constitute a potential novel treatment strategy for panic and related anxiety disorders. The neural pathways through which ORX1 receptor antagonists attenuate panic responses involve the extended amygdala, periaqueductal gray, and medullary autonomic centers.


Assuntos
Encéfalo/efeitos dos fármacos , Pânico/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Neuropeptídeos/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Encéfalo/fisiologia , Cafeína/farmacologia , Carbolinas/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Antagonistas GABAérgicos/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Masculino , Bulbo/efeitos dos fármacos , Bulbo/fisiologia , Receptores de Orexina , Pânico/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/fisiologia
16.
Neuropsychopharmacology ; 37(9): 1999-2011, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22569505

RESUMO

Orexins (OX) and their receptors (OXR) modulate feeding, arousal, stress, and drug abuse. Neural systems that motivate and reinforce drug abuse may also underlie compulsive food seeking and intake. Therefore, the effects of GSK1059865 (5-bromo-N-[(2S,5S)-1-(3-fluoro-2-methoxybenzoyl)-5-methylpiperidin-2-yl]methyl-pyridin-2-amine), a selective OX(1)R antagonist, JNJ-10397049 (N-(2,4-dibromophenyl)-N'-[(4S,5S)-2,2-dimethyl-4-phenyl-1,3-dioxan-5-yl]urea), a selective OX(2)R antagonist, and SB-649868 (N-[((2S)-1-{[5-(4-fluorophenyl)-2-methyl-1,3-thiazol-4-yl]carbonyl}-2-piperidinyl)methyl]-1-benzofuran-4-carboxamide), a dual OX(1)/OX(2)R antagonist were evaluated in a binge eating (BE) model in female rats. BE of highly palatable food (HPF) was evoked by three cycles of food restriction followed by stress, elicited by exposing rats to HPF, but preventing them from having access to it for 15 min. Pharmacokinetic assessments of all compounds were obtained under the same experimental conditions used for the behavioral experiments. Topiramate was used as the reference compound as it selectively blocks BE in rats and humans. Dose-related thresholds for sleep-inducing effects of the OXR antagonists were measured using polysomnography in parallel experiments. SB-649868 and GSK1059865, but not JNJ-10397049, selectively reduced BE for HPF without affecting standard food pellet intake, at doses that did not induce sleep. These results indicate, for the first time, a major role of OX(1)R mechanisms in BE, suggesting that selective antagonism at OX(1)R could represent a novel pharmacological treatment for BE and possibly other eating disorders with a compulsive component.


Assuntos
Bulimia/metabolismo , Comportamento Compulsivo , Ingestão de Alimentos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Animais , Bulimia/tratamento farmacológico , Bulimia/psicologia , Comportamento Compulsivo/tratamento farmacológico , Comportamento Compulsivo/psicologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/psicologia , Feminino , Frutose/análogos & derivados , Frutose/farmacologia , Frutose/uso terapêutico , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Masculino , Neuropeptídeos/farmacologia , Receptores de Orexina , Orexinas , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/antagonistas & inibidores , Esquema de Reforço , Fatores Sexuais , Topiramato , Células Tumorais Cultivadas
17.
Neuropsychopharmacology ; 37(8): 1911-22, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22453138

RESUMO

Acute hypercapnia (elevated arterial CO(2)/H(+)) is a suffocation signal that is life threatening and rapidly mobilizes adaptive changes in breathing and behavioral arousal in order to restore acid-base homeostasis. Severe hypercapnia, seen in respiratory disorders (eg, asthma or bronchitis, chronic obstructive pulmonary disease (COPD)), also results in high anxiety and autonomic activation. Recent evidence has demonstrated that wake-promoting hypothalamic orexin (ORX: also known as hypocretin) neurons are highly sensitive to local changes in CO(2)/H(+), and mice lacking prepro-ORX have blunted respiratory responses to hypercapnia. Furthermore, in a recent clinical study, ORX-A, which crosses blood-brain barrier easily, was dramatically increased in the plasma of patients with COPD and hypercapnic respiratory failure. This is consistent with a rodent model of COPD where chronic exposure to cigarette smoke led to a threefold increase in hypothalamic ORX-A expression. In the present study, we determined the role of ORX in the anxiety-like behavior and cardiorespiratory responses to acute exposure to a threshold panic challenge (ie, 20% CO(2)/normoxic gas). Exposing conscious rats to such hypercapnic, but not atmospheric air, resulted in respiratory, pressor, and bradycardic responses, as well as anxiety-like behavior and increased cellular c-Fos responses in ORX neurons. Systemically, pre-treating rats with a centrally active ORX1 receptor antagonist (30 mg/kg SB334867) attenuated hypercapnic gas-induced pressor and anxiety responses, without altering the robust bradycardia response, and only attenuated breathing responses at offset of the CO(2) challenge. Our results show that the ORX system has an important role in anxiety and sympathetic mobilization during hypercapnia. Furthermore, ORX1 receptor antagonists may be a therapeutic option rapidly treating increased anxiety and sympathetic drive seen during panic attacks and in hypercapnic states such as COPD.


Assuntos
Ansiedade/fisiopatologia , Bradicardia/fisiopatologia , Hipercapnia/fisiopatologia , Hipertensão/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Animais , Ansiedade/induzido quimicamente , Ansiedade/complicações , Benzoxazóis/farmacologia , Bradicardia/induzido quimicamente , Bradicardia/complicações , Modelos Animais de Doenças , Hipercapnia/induzido quimicamente , Hipercapnia/complicações , Hipertensão/induzido quimicamente , Hipertensão/complicações , Masculino , Naftiridinas , Neurônios/fisiologia , Receptores de Orexina , Orexinas , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Respiração/efeitos dos fármacos , Ureia/análogos & derivados , Ureia/farmacologia
18.
Pflugers Arch ; 463(4): 531-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22350729

RESUMO

The effects of the centrally administered neuropeptides orexin-A on water intake and vasopressin (VP) secretion were studied in male Wistar rats (180-250 g). Different doses (10, 30, and 90 µg/10 µl) of the orexins and the specific orexin receptor-1 (OX(1)) antagonist SB 408124 (30 µg/10 µl) were administered intracerebroventricularly (i.c.v.) under anaesthesia, and the water consumption was measured during 6 h. A plasma VP level elevation was induced by histamine (10 mg/kg) or 2.5% NaCl (10 ml/kg) administered intraperitoneally (i.p.). The plasma VP levels were measured by radioimmunoassay. Increased water consumption was observed after the administration of 30 µg/10 µl orexin-A. There were no changes in basal VP secretion after the administration of different doses of the orexins. A significant increase in plasma VP concentration was detected following histamine administration. After 2.5% NaCl administration, there was a moderate VP level enhancement. Intracerebroventricularly administered orexin-A (30 µg/10 µl) blocked the VP level increase induced by either histamine or 2.5% NaCl administration. The inhibitory effects were prevented by the specific OX(1) receptor antagonist. In conclusion, the orexins increased water consumption. After 30 µg/10 µl orexin-A administration, the polydipsia was more pronounced. The OX(1) receptor antagonist significantly decreased the polydipsia. Histamine or hyperosmotic VP release enhancement was blocked by previously administered orexin. This inhibition was not observed following OX(1) receptor antagonist administration. Our results suggest that the effects of the orexins on water consumption or blockade of the histamine and osmosis-induced VP level increase are mediated by the OX(1) receptor.


Assuntos
Histamina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Neuropeptídeos/administração & dosagem , Vasopressinas/sangue , Animais , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/fisiologia , Histamina/farmacologia , Injeções Intraventriculares , Masculino , Neurotransmissores/administração & dosagem , Receptores de Orexina , Orexinas , Pressão Osmótica , Compostos de Fenilureia/farmacologia , Polidipsia/induzido quimicamente , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/antagonistas & inibidores , Receptores de Neuropeptídeos/fisiologia , Vasopressinas/metabolismo
19.
J Mol Neurosci ; 46(1): 10-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21547533

RESUMO

Orexins A and B are newly discovered neuropeptides with pleiotropic activity. They signal through two G protein-coupled receptors: OX(1) and OX(2). In this study, we examined the expression of orexin receptors and effects of the receptors' activation on cyclic AMP formation in the primary neuronal cell cultures from rat cerebral cortex. Both types of orexin receptors were expressed in rat cortical neurons; the level of OX(2)R was markedly higher compared to OX(1)R. Orexin A (an agonist of OX(1)R and OX(2)R) and [Ala(11)-D-Leu(15)]orexin B (a selective agonist of OX(2)R) did not affect basal cyclic AMP formation in the primary neuronal cell cultures. Both peptides (0.001-1 µM) inhibited, in a concentration-dependent manner and IC(50) values in low nanomolar range, the increase in the nucleotide production evoked by forskolin (1 µM; a direct activator of adenylyl cyclase), pituitary adenylate cyclase-activating polypeptide (PACAP27; 0.1 µM), and vasoactive intestinal peptide (VIP; 3 µM). Effects of orexin A on forskolin-, PACAP27-, and VIP-stimulated cyclic AMP synthesis were blocked by TCS OX2 29 (a selective antagonist of OX(2)R), and unaffected by SB 408124 (a selective antagonist of OX(1)R). Pretreatment of neuronal cell cultures with pertussis toxin (PTX) abolished the inhibitory action of orexin A on forskolin- and PACAP-stimulated cyclic AMP accumulation. It is suggested that in cultured rat cortical neurons orexins, acting at OX(2) receptors coupled to PTX-sensitive G(i) protein, inhibit cyclic AMP synthesis.


Assuntos
AMP Cíclico/antagonistas & inibidores , AMP Cíclico/biossíntese , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Animais , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neurônios/citologia , Neuropeptídeos/fisiologia , Receptores de Orexina , Orexinas , Cultura Primária de Células , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
Biol Psychiatry ; 71(3): 214-23, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21831361

RESUMO

BACKGROUND: Hypocretin (orexin) signaling is involved in drug addiction. In this study, we investigated the role of these hypothalamic neuropeptides in nicotine withdrawal by using behavioral and neuroanatomical approaches. METHODS: Nicotine withdrawal syndrome was precipitated by mecamylamine (2 mg/kg, subcutaneous) in C57BL/6J nicotine-dependent mice (25 mg/kg/day for 14 days) pretreated with the hypocretin receptor 1 (Hcrtr-1) antagonist SB334867 (5 and 10 mg/kg, intraperitoneal), the hypocretin receptor 2 antagonist TCSOX229 (5 and 10 mg/kg, intraperitoneal), and in preprohypocretin knockout mice. c-Fos expression was analyzed in several brain areas related to nicotine dependence by immunofluorescence techniques. Retrograde tracing with rhodamine-labeled fluorescent latex microspheres was used to determine whether the hypocretin neurons project directly to the paraventricular nucleus of the hypothalamus (PVN), and SB334867 was locally administered intra-PVN (10 nmol/side) to test the specific involvement of Hcrtr-1 in this brain area during nicotine withdrawal. RESULTS: Somatic signs of nicotine withdrawal were attenuated in mice pretreated with SB334867 and in preprohypocretin knockout mice. No changes were found in TCSOX229 pretreated animals. Nicotine withdrawal increased the percentage of hypocretin cells expressing c-Fos in the perifornical, dorsomedial, and lateral hypothalamus. In addition, the increased c-Fos expression in the PVN during withdrawal was dependent on hypocretin transmission through Hcrtr-1 activation. Hypocretin neurons directly innervate the PVN and the local infusion of SB334867 into the PVN decreased the expression of nicotine withdrawal. CONCLUSIONS: These data demonstrate that hypocretin signaling acting on Hcrtr-1 in the PVN plays a crucial role in the expression of nicotine withdrawal.


Assuntos
Antígenos de Superfície/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Tabagismo/fisiopatologia , Animais , Benzoxazóis/administração & dosagem , Benzoxazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoquinolinas/farmacologia , Masculino , Mecamilamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções , Imagem Molecular/métodos , Naftiridinas , Técnicas de Rastreamento Neuroanatômico/métodos , Neuropeptídeos/genética , Receptores de Orexina , Orexinas , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Piridinas/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Ureia/administração & dosagem , Ureia/análogos & derivados , Ureia/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA