Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Res ; 1680: 155-164, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29274880

RESUMO

Mouse VD-hemopressin(α) (VD-Hpα) is an undecapeptide that selectively activates CB1 cannabinoid receptor in in vitro functional tests, and exerts CB1-mediated central antinociception in the mouse tail-flick assay. The aim of the present study was to further investigate the analgesic properties of supraspinal mouse VD-Hpα in a range of preclinical pain models. Our results indicated that the classical cannabinoid agonist WIN 55,212-2 produced supraspinal analgesia in preclinical pain models, which was selectively antagonized by the CB1 antagonist/inverse agonist AM251, but not by the CB2 antagonist AM630. In contrast, in post-operative pain model and phase I of formalin test, intracerebroventricular administration of mouse VD-Hpα induced dose-related analgesia in mice, which were markedly reduced by pretreatment with the CB1 neutral antagonist AM4113, but not AM251, AM630 and the selective antagonists of opioid and Transient Receptor Potential Vanilloid Type 1 (TRPV1) receptors. Furthermore, in the acetic acid-induced visceral pain model, supraspinal administration of mouse VD-Hpα dose-dependently produced analgesic activities and the effects were significantly antagonized by both AM4113 and the TRPV1 receptor antagonist SB366791, but not AM251, AM630 and naloxone. In addition, central injection of mouse VD-Hpα did not have significant effect in phase II of formalin test. Taken together, the present work suggests that the CB1 receptor peptidic agonist mouse VD-Hpα produces supraspinal analgesia in preclinical pain models via a novel CB1 receptor-mediated mechanism, in a manner pharmacologically dissociable from WIN 55,212-2. In addition, TRPV1 receptor might also be involved in mouse VD-Hpα-induced analgesia in a visceral pain model.


Assuntos
Analgésicos/uso terapêutico , Agonistas de Receptores de Canabinoides/uso terapêutico , Oligopeptídeos/uso terapêutico , Dor/tratamento farmacológico , Ácido Acético/toxicidade , Animais , Benzoxazinas/uso terapêutico , Antagonistas de Receptores de Canabinoides/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Formaldeído/toxicidade , Injeções Espinhais , Masculino , Camundongos , Morfolinas/uso terapêutico , Naftalenos/uso terapêutico , Dor/etiologia , Medição da Dor , Teste de Desempenho do Rota-Rod
2.
J Med Chem ; 59(22): 10198-10208, 2016 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-27798836

RESUMO

The opioid and neuropeptide FF pharmacophore-containing chimeric peptide 0 (BN-9) was recently developed and produced potent nontolerance forming analgesia. In this study, 11 analogues of 0 were designed and synthesized. An in vitro cAMP assay demonstrated that these analogues behaved as multifunctional agonists at both opioid and NPFF receptors. In mouse tail-flick test, most of the analogues produced potent nontolerance forming antinociception. Notably, 11 (DN-9) was 33-fold more potent than 0 at analgesic effects, which was mediated by µ- and κ-opioid receptors. In addition, 11 also produced powerful analgesic effects in the formalin pain and CFA-induced chronic inflammatory pain models. Strikingly, following its repeated administration for 6 days, 11 did not produce antinociceptive tolerance in the tail-flick test and CFA-induced pain model. The present work indicates that it is reasonable to design multifunctional peptide ligands for opioid and NPFF receptors in a single molecule producing effective nontolerance forming antinociception.


Assuntos
Analgésicos/farmacologia , Dor/tratamento farmacológico , Peptídeos/farmacologia , Receptores de Neuropeptídeos/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Analgésicos/síntese química , Analgésicos/química , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Relação Estrutura-Atividade
3.
Peptides ; 86: 24-32, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27669639

RESUMO

Neurite outgrowth is an important process in neural regeneration and plasticity, especially after neural injury, and recent evidence indicates that several Gαi/o protein-coupled receptors play an important role in neurite outgrowth. The neuropeptide (NP)FF system contains two Gαi/o protein-coupled receptors, NPFF1 and NPFF2 receptors, which are mainly distributed in the central nervous system. The aim of the present study was to determine whether the NPFF system is involved in neurite outgrowth in Neuro 2A cells. We showed that Neuro 2A cells endogenously expressed NPFF2 receptor, and the NPFF2 receptor agonist dNPA inhibited cyclic adenosine monophosphate (cAMP) production stimulated by forskolin in Neuro 2A cells. We also demonstrated that NPFF and dNPA dose-dependently induced neurite outgrowth in Neuro 2A cells, which was completely abolished by the NPFF receptor antagonist RF9. Pretreatment with mitogen-activated protein kinase inhibitors PD98059 and U0126 decreased dNPA-induced neurite outgrowth. In addition, dNPA increased phosphorylation of extracellular signal-regulated kinase (ERK) in Neuro 2A cells, which was completely antagonized by pretreatment with U0126. Our results suggest that activation of NPFF2 receptor stimulates neurite outgrowth in Neuro 2A cells through activation of the ERK signaling pathway. Moreover, NPFF2 receptor may be a potential therapeutic target for neural injury and degeneration in the future.


Assuntos
Sistema de Sinalização das MAP Quinases , Neuritos/fisiologia , Receptores de Neuropeptídeos/metabolismo , Animais , Crescimento Celular , Linhagem Celular , Colforsina/metabolismo , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Camundongos , Oligopeptídeos/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/genética
4.
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
5.
Eur J Pharmacol ; 767: 119-25, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26472125

RESUMO

Neuropeptide FF (NPFF) system has recently been reported to modulate cannabinoid-induced antinociception. The aim of the present study was to further investigate the roles of NPFF system in the antinociceptive effects induced by intracerebroventricular (i.c.v.) administration of mouse VD-hemopressin(α), a novel endogenous agonist of cannabinoid CB1 receptor, in naive and VD-hemopressin(α)-tolerant mice. The effects of NPFF system on the antinociception induced by VD-hemopressin(α) were investigated in the radiant heat tail-flick test in naive mice and VD-hemopressin(α)-tolerant mice. The cannabinoid-tolerant mice were produced by given daily injections of VD-hemopressin(α) (20 nmol, i.c.v.) for 5 days and the antinociception was measured on day 6. In naive mice, intracerebroventricular injection of NPFF dose-dependently attenuated central analgesia of VD-hemopressin(α). In contrast, neuropeptide VF (NPVF) and D.NP(N-Me)AFLFQPQRF-NH2 (dNPA), two highly selective agonists for Neuropeptide FF1 and Neuropeptide FF2 receptors, enhanced VD-hemopressin(α)-induced antinociception in a dose-dependent manner. In addition, the VD-hemopressin(α)-modulating activities of NPFF and related peptides were antagonized by the Neuropeptide FF receptors selective antagonist 1-adamantanecarbonyl-RF-NH2 (RF9). In VD-hemopressin(α)-tolerant mice, NPFF failed to modify VD-hemopressin(α)-induced antinociception. However, both neuropeptide VF and dNPA dose-dependently potentiated the antinociception of VD-hemopressin(α) and these cannabinoid-potentiating effects were reduced by RF9. The present works support the cannabinoid-modulating character of NPFF system in naive and cannabinoid-tolerant mice. In addition, the data suggest that a chronic cannabinoid treatment modifies the pharmacological profiles of NPFF, but not the cannabinoid-potentiating effects of neuropeptide VF and dNPA.


Assuntos
Analgésicos/farmacologia , Neuropeptídeos/farmacologia , Oligopeptídeos/farmacologia , Dor/tratamento farmacológico , Adamantano/análogos & derivados , Adamantano/farmacologia , Analgésicos/administração & dosagem , Animais , Dipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Infusões Intraventriculares , Masculino , Camundongos , Neuropeptídeos/administração & dosagem , Neuropeptídeos/antagonistas & inibidores , Oligopeptídeos/administração & dosagem , Oligopeptídeos/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/antagonistas & inibidores
6.
PLoS One ; 9(8): e103773, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25090615

RESUMO

Neuropeptide FF (NPFF) is known to be an endogenous opioid-modulating peptide. Nevertheless, very few researches focused on the interaction between NPFF and endogenous opioid peptides. In the present study, we have investigated the effects of NPFF system on the supraspinal antinociceptive effects induced by the endogenous µ-opioid receptor agonists, endomorphin-1 (EM-1) and endomorphin-2 (EM-2). In the mouse tail-flick assay, intracerebroventricular injection of EM-1 induced antinociception via µ-opioid receptor while the antinociception of intracerebroventricular injected EM-2 was mediated by both µ- and κ-opioid receptors. In addition, central administration of NPFF significantly reduced EM-1-induced central antinociception, but enhanced EM-2-induced central antinociception. The results using the selective NPFF1 and NPFF2 receptor agonists indicated that the EM-1-modulating action of NPFF was mainly mediated by NPFF2 receptor, while NPFF potentiated EM-2-induecd antinociception via both NPFF1 and NPFF2 receptors. To further investigate the roles of µ- and κ-opioid systems in the opposite effects of NPFF on central antinociception of endomprphins, the µ- and κ-opioid receptors selective agonists DAMGO and U69593, respectively, were used. Our results showed that NPFF could reduce the central antinociception of DAMGO via NPFF2 receptor and enhance the central antinociception of U69593 via both NPFF1 and NPFF2 receptors. Taken together, our data demonstrate that NPFF exerts opposite effects on central antinociception of endomorphins and provide the first evidence that NPFF potentiate antinociception of EM-2, which might result from the interaction between NPFF and κ-opioid systems.


Assuntos
Nociceptividade/efeitos dos fármacos , Oligopeptídeos/farmacologia , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Benzenoacetamidas/farmacologia , Dipeptídeos/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Pirrolidinas/farmacologia
7.
Neurosci Lett ; 578: 187-91, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25010019

RESUMO

The nonapeptide hemopressin and its N-terminal extension VD-hemopressin(α) were reported as an antagonist/inverse agonist and an agonist of CB1 receptor, respectively. These novel cannabinoid peptides have been demonstrated to modulate the acute pain. In the present study, hemopressin (11, 22 and 45 nmol, i.c.v.) dose-dependently produced antinociception after supraspinal administration in the radiant heat tail-flick test. Furthermore, the development of antinociceptive tolerance to hemopressin, VD-hemopressin(α) and WIN55,212-2, and cross-tolerance among these cannabinoids were investigated in mice. The tolerance developed on day 4 after supraspinal injection of hemopressin (45 nmol), VD-hemopressin(α) (20 nmol) and WIN55,212-2 (7.5 nmol). Our results indicated symmetrical cross-tolerance between hemopressin, VD-hemopressin(α) and WIN55,212-2 at the supraspinal level in mice. These results demonstrate that both hemopressin and VD-hemopressin(α) have a time-course and extent of tolerance similar to the synthetic cannabinoid WIN55,212-2. In addition, our data imply that a common mechanism is involved in the antinociception of the three cannabinoid ligands.


Assuntos
Analgésicos/farmacologia , Benzoxazinas/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Hemoglobinas/farmacologia , Morfolinas/farmacologia , Naftalenos/farmacologia , Nociceptividade/efeitos dos fármacos , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Agonismo Inverso de Drogas , Tolerância a Medicamentos , Ligantes , Masculino , Camundongos , Medula Espinal , Cauda
8.
Peptides ; 56: 45-51, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24681436

RESUMO

Previous studies suggest that cannabinoids system plays an important role in cardiovascular regulation. (m)VD-hemopressin(α) (VD-Hpα), an 11-residue peptide originating from the α1 chain of hemoglobin, was recently reported as a selective agonist of cannabinoid CB1 receptor. The present study was undertaken to investigate the intrathecal (i.t.) action of (m)VD-Hpα on blood pressure in urethane-anesthetized rats. Our results demonstrated that injections of (m)VD-Hpα (5-30 nmol, i.t.) produced a dose-dependent decrease in mean arterial pressure (MAP), similar to that of the non-peptidic cannabinoid receptor agonist WIN55212-2 (1.25-10 nmol, i.t.). The hypotensive effect of (m)VD-Hpα was not influenced by the CB1 receptor antagonist AM251 (20 nmol, i.t.) or the CB2 receptor antagonist AM630 (20 nmol, i.t.). However, WIN55212-2-induced hypotension was almost completely prevented by i.t. administration of AM251, not by AM630. The spinal hypotension of (m)VD-Hpα and WIN55212-2 was significantly reduced by pretreatment with the α-adrenoceptor antagonist phentolamine (1 mg/kg, i.v.), but not by the ß-adrenoceptor antagonist propranolol (2 mg/kg, i.v.) or the muscarinic receptor antagonist atropine (2 mg/kg, i.v.). In addition, L-NAME (50 mg/kg, i.v.), the inhibitor of nitric oxide (NO) synthase, significantly reduced WIN55212-2-induced hypotension, but had no effect on the hypotensive response to (m)VD-Hpα. Collectively, the results show that i.t. administration of (m)VD-Hpα induces a decrease in MAP via a non-CB1 and non-CB2 mechanism.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hemoglobinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Uretana/farmacologia , Animais , Hemoglobinas/administração & dosagem , Humanos , Hipotensão/induzido quimicamente , Injeções Espinhais , Masculino , Óxido Nítrico/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Wistar , Receptor CB2 de Canabinoide/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...