Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 613(1-3): 54-9, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19386271

RESUMO

Despite the clear roles played by peroxisome proliferators-activated receptor alpha (PPAR-alpha) in lipid metabolism, inflammation and feeding, the effects of its activation in the central nervous system (CNS) are largely unknown. Palmitoylethanolamide (PEA), a member of the fatty-acid ethanolamide family, acts peripherally as an endogenous PPAR-alpha agonist, exerting analgesic and anti-inflammatory effects. Both PPAR-alpha and PEA are present in the CNS, but the specific functions of this lipid and its receptor remain to be clarified. Using the carrageenan-induced paw model of hyperalgesia in mice, we report here that intracerebroventricular administration of PEA (0.1-1 microg) 30 min before carrageenan injection markedly reduced mechanical hyperalgesia up to 24 h following inflammatory insult. This effect was mimicked by GW7647 (1 microg), a synthetic PPAR-alpha agonist. The obligatory role of PPAR-alpha in mediating PEA's actions was confirmed by the lack of anti-hyperalgesic effects in mutant mice lacking PPAR-alpha. PEA significantly reduced the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in sciatic nerves and restored carrageenan-induced reductions of PPAR-alpha in the L4-L6 dorsal root ganglia (DRG). To investigate the mechanism by which PEA attenuated hyperalgesia, we evaluated inhibitory kB-alpha (IkB-alpha) degradation and p65 nuclear factor kB (NF-kappaB) activation in DRG. PEA prevented IkB-alpha degradation and p65 NF-kappaB nuclear translocation, confirming the involvement of this transcriptional factor in the control of peripheral hyperalgesia. These results add further support to the broad-spectrum of biological and pharmacological effects induced by PPAR-alpha agonists, suggesting a centrally mediated component for these drugs in controlling inflammatory pain.


Assuntos
Núcleo Celular/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/patologia , NF-kappa B/metabolismo , Ácidos Palmíticos/administração & dosagem , Ácidos Palmíticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Amidas , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Butiratos/administração & dosagem , Butiratos/farmacologia , Carragenina/toxicidade , Núcleo Celular/metabolismo , Sistema Nervoso Central/metabolismo , Ciclo-Oxigenase 2/biossíntese , Endocanabinoides , Indução Enzimática/efeitos dos fármacos , Etanolaminas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Camundongos , Óxido Nítrico Sintase Tipo II/biossíntese , PPAR alfa/agonistas , PPAR alfa/metabolismo , Ácidos Palmíticos/uso terapêutico , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/farmacologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo
2.
Bioorg Med Chem Lett ; 19(3): 639-43, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19128970

RESUMO

Cannabinoid CB(1) receptor antagonists reduce body weight in rodents and humans, but their clinical utility as anti-obesity agents is limited by centrally mediated side effects. Here, we describe the first mixed CB(1) antagonist/CB(2) agonist, URB447 ([4-amino-1-(4-chlorobenzyl)-2-methyl-5-phenyl-1H-pyrrol-3-yl](phenyl)methanone), which lowers food intake and body-weight gain in mice without entering the brain or antagonizing central CB(1)-dependent responses. URB447 may provide a useful pharmacological tool for investigating the cannabinoid system, and might serve as a starting point for developing clinically viable CB(1) antagonists devoid of central side effects.


Assuntos
Compostos de Benzil/síntese química , Canabinoides/antagonistas & inibidores , Canabinoides/química , Química Farmacêutica/métodos , Pirróis/síntese química , Animais , Fármacos Antiobesidade/farmacologia , Compostos de Benzil/farmacologia , Peso Corporal , Cerebelo/metabolismo , Desenho de Fármacos , Comportamento Alimentar/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Químicos , Pirróis/farmacologia , Ratos , Receptores de Canabinoides/metabolismo , Aumento de Peso/efeitos dos fármacos
3.
Brain Res Rev ; 60(1): 114-24, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19162071

RESUMO

We discuss and present new data regarding the physiological and molecular mechanisms of nuclear receptor activation in pain control, with a particular emphasis on non-genomic effects of ligands at peroxisome proliferator-activated receptor (PPAR), GPR30, and classical estrogen receptors. PPARalpha agonists rapidly reduce both acute and chronic pain in a number of pain assays. These effects precede transcriptional anti-inflammatory actions, and are mediated in part by IK(ca) and BK(ca) channels on DRG neurons. In contrast to the peripheral site of action of PPARalpha ligands, the dorsal horn supports the expression of PPARgamma. Intrathecal administration of PPARgamma ligands rapidly (< or =5 min) attenuated mechanical and thermal hypersensitivity associated with nerve injury in a dose-dependent manner that could be blocked with PPARgamma antagonists. By contrast, a PPARgamma antagonist itself rapidly increased the mechanical allodynia associated with nerve injury. These data suggest that ligand-dependent, non-genomic activation of spinal PPARgamma decreases behavioral signs of inflammatory and neuropathic pain. We also report that the GPR30 is expressed on cultured sensory neurons, that activation of the receptor elicits signaling to increase calcium accumulation. This signaling may contribute to increased neuronal sensitivity as treatment with the GPR30 agonist induces hyperalgesia. Finally, application of the membrane-impermeable 17beta-E(2)-BSA rapidly (within 15 min) enhanced BK-stimulated inositol phosphate (IP) accumulation and PGE(2)-mediated cAMP accumulation in trigeminal ganglion cultures. We conclude that nuclear receptor ligands may operate through rapid, non-genomic mechanisms to modulate inflammatory and neuropathic pain.


Assuntos
Analgésicos/farmacologia , Nociceptores/efeitos dos fármacos , Dor/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Humanos , Nociceptores/metabolismo , PPAR alfa/efeitos dos fármacos , PPAR alfa/metabolismo , PPAR gama/efeitos dos fármacos , PPAR gama/metabolismo , Dor/metabolismo , Dor/fisiopatologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Células Receptoras Sensoriais/metabolismo
4.
Anesth Analg ; 104(6): 1563-9, table of contents, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17513659

RESUMO

BACKGROUND: Propofol can inhibit fatty acid amidohydrolase, the enzyme responsible for the metabolism of anandamide (an endocannabinoid). To study the potential antinociceptive effect of propofol, we administered different doses (0.005, 0.05, 0.5, 5, and 500 microg) of the anesthetic in the hind paw of animals to determine an ED50. To further investigate the mechanisms by which propofol produced its antinociceptive effect, we used specific antagonists for the cannabinoid CB1 (AM251) and CB2 (AM630) receptors and measured fatty-acid amide/endocannabinoid (anandamide, 2-arachidonylglycerol, and palmitoylethanolamide) concentrations in skin paw tissues. METHODS: Formalin tests were performed on 65 Wistar rats allocated to six different groups: 1) control (Intralipidtrade mark 10%); 2) propofol (ED50 dose); 3) AM251; 4) AM251 + propofol; 5) AM630; 6) AM630 + propofol. Drugs were injected subcutaneously in the dorsal surface of the hind paw (50 microL) 15 min before 2.5% formalin injection into the same paw. Fatty-acid amide/endocannabinoid levels were measured by high performance liquid chromatography/mass spectrometry analysis. RESULTS: Propofol produced a dose-dependent antinociceptive effect for the early and late phases of the formalin test with an ED50 of 0.08 +/- 0.061 microg for the latter phase. This effect was antagonized by AM251 and AM630. It was locally mediated, since a higher dose of propofol given in the contralateral paw was not antinociceptive. Finally, only paw concentrations of palmitoylethanolamide were significantly increased. CONCLUSION: In a test of inflammatory pain, locally injected propofol decreased pain behavior in a dose-dependent manner. This antinociceptive effect was mediated, in part, by CB1 and CB2 receptors.


Assuntos
Analgésicos/administração & dosagem , Moduladores de Receptores de Canabinoides/fisiologia , Propofol/administração & dosagem , Receptor CB1 de Canabinoide/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Injeções Subcutâneas , Masculino , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Wistar
5.
Eur J Pharmacol ; 566(1-3): 117-9, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17434479

RESUMO

The analgesic properties of cannabinoid receptor agonists are well characterized. However, numerous side effects limit the therapeutic potential of these agents. Here we report a synergistic antinociceptive interaction between the endogenous cannabinoid receptor agonist anandamide and the synthetic peroxisome proliferator-activated receptor-alpha (PPAR-alpha) agonist 2-(4-(2-(1-Cyclohexanebutyl)-3-cyclohexylureido)ethyl)phenylthio)-2-methylpropionic acid (GW7647) in a model of acute chemical-induced pain. Moreover, we show that anandamide synergistically interacts with the large-conductance potassium channel (KCa1.1, BK) activator isopimaric acid. These findings reveal a synergistic interaction between the endocannabinoid and PPAR-alpha systems that might be exploited clinically and identify a new pharmacological effect of the BK channel activator isopimaric acid.


Assuntos
Analgésicos/farmacologia , Ácidos Araquidônicos/farmacologia , Butiratos/farmacologia , Agonistas de Receptores de Canabinoides , PPAR alfa/agonistas , Compostos de Fenilureia/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Sinergismo Farmacológico , Endocanabinoides , Formaldeído , Masculino , Camundongos , Dor/induzido quimicamente , Dor/tratamento farmacológico , Dor/fisiopatologia
6.
J Pharmacol Exp Ther ; 322(1): 236-42, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412883

RESUMO

Fatty acid amide hydrolase (FAAH) is an intracellular serine hydrolase that catalyzes the cleavage of bioactive fatty acid ethanolamides, such as the endogenous cannabinoid agonist anandamide. Genetic deletion of the faah gene in mice elevates brain anandamide levels and amplifies the antinociceptive effects of this compound. Likewise, pharmacological blockade of FAAH activity reduces nocifensive behavior in animal models of acute and inflammatory pain. In the present study, we investigated the effects of the selective FAAH inhibitor URB597 (KDS-4103, cyclohexylcarbamic acid 3'-carbamoylbiphenyl-3-yl ester) in the mouse chronic constriction injury (CCI) model of neuropathic pain. Oral administration of URB597 (1-50 mg/kg, once daily) for 4 days produced a dose-dependent reduction in nocifensive responses to thermal and mechanical stimuli, which was prevented by a single i.p. administration of the cannabinoid CB(1) receptor antagonist rimonabant (1 mg/kg). The antihyperalgesic effects of URB597 were accompanied by a reduction in plasma extravasation induced by CCI, which was prevented by rimonabant (1 mg/kg i.p.) and attenuated by the CB(2) antagonist SR144528 (1 mg/kg i.p.). Oral dosing with URB597 achieved significant, albeit transient, drug levels in plasma, inhibited brain FAAH activity, and elevated spinal cord anandamide content. The results provide new evidence for a role of the endocannabinoid system in pain modulation and reinforce the proposed role of FAAH as a target for analgesic drug development.


Assuntos
Amidoidrolases/antagonistas & inibidores , Analgésicos/farmacologia , Benzamidas/farmacologia , Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Hiperalgesia/tratamento farmacológico , Administração Oral , Animais , Benzamidas/administração & dosagem , Permeabilidade Capilar/efeitos dos fármacos , Carbamatos/administração & dosagem , Masculino , Camundongos , Receptor CB1 de Canabinoide/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Medula Espinal/química , Medula Espinal/efeitos dos fármacos
7.
Eur J Pharmacol ; 550(1-3): 68-77, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17027744

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit fatty-acid amide hydrolase (FAAH), the enzyme responsible for the metabolism of anandamide, an endocannabinoid. It has been suggested that the mechanisms of action of NSAIDs could be due to inhibition of cyclooxygenase (COX) and also to an increase in endocannabinoid concentrations. In a previous study we have demonstrated that the local analgesic interaction between anandamide and ibuprofen (a non-specific COX inhibitor) was synergistic for the acute and inflammatory phases of the formalin test. To test this hypothesis further, we repeated similar experiments with rofecoxib (a selective COX-2 inhibitor) and also measured the local concentrations of anandamide, and of two fatty-acid amides, oleoylethanolamide and palmitoylethanolamide. We established the ED(50) for anandamide (34.52 pmol+/-17.26) and rofecoxib (381.72 pmol+/-190.86) and showed that the analgesic effect of the combination was synergistic. We also found that paw tissue levels of anandamide, oleoylethanolamide and palmitoylethanolamide were significantly higher when anandamide was combined with NSAIDs and that this effect was greater with rofecoxib. In conclusion, local injection of anandamide or rofecoxib was antinociceptive in a test of acute and inflammatory pain and the combination of anandamide with rofecoxib was synergistic. Finally, locally injected anandamide with either NSAID (ibuprofen or rofecoxib) generates higher amount of fatty-acid ethanolamides. The exact comprehension of the mechanisms involved needs further investigation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Ácidos Araquidônicos/farmacologia , Sistema Nervoso Periférico/efeitos dos fármacos , Alcamidas Poli-Insaturadas/farmacologia , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Cromatografia Líquida de Alta Pressão , Inibidores de Ciclo-Oxigenase 2/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Edema/induzido quimicamente , Edema/tratamento farmacológico , Endocanabinoides , Formaldeído , Ibuprofeno/farmacologia , Lactonas/farmacologia , Masculino , Espectrometria de Massas , Nitrobenzenos/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Wistar , Sulfonamidas/farmacologia , Sulfonas/farmacologia , Canais de Cátion TRPV/efeitos dos fármacos
8.
J Pharmacol Exp Ther ; 319(3): 1051-61, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16997973

RESUMO

Severe pain remains a major area of unmet medical need. Here we report that agonists of the nuclear receptor PPAR-alpha (peroxisome proliferator-activated receptor-alpha) suppress pain behaviors induced in mice by chemical tissue injury, nerve damage, or inflammation. The PPAR-alpha agonists GW7647 [2-(4-(2-(1-cyclohexanebutyl)-3-cyclohexylureido)ethyl)phenylthio)-2-methylpropionic acid], Wy-14643 [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid], and palmitoylethanolamide (PEA) reduced nocifensive behaviors elicited in mice by intraplantar (i.pl.) injection of formalin or i.p. injection of magnesium sulfate. These effects were absent in PPAR-alpha-null mice yet occurred within minutes of agonist administration in wild-type mice, suggesting that they were mediated through a transcription-independent mechanism. Consistent with this hypothesis, blockade of calcium-operated IK(ca) (K(Ca)3.1) and BK(ca) (K(Ca)1.1) potassium channels prevented the effects of GW7647 and PEA in the formalin test. Three observations suggest that PPAR-alpha agonists may inhibit nocifensive responses by acting on peripheral PPAR-alpha. (i) PEA reduced formalin-induced pain at i.pl. doses that produced no increase in systemic PEA levels; (ii) PPAR-alpha was expressed in dorsal root ganglia neurons of wild-type but not PPAR-alpha-null mice; and (ii) GW7647 and PEA prevented formalin-induced firing of spinal cord nociceptive neurons in rats. In addition to modulating nociception, GW7647 and PEA reduced hyperalgesic responses in the chronic constriction injury model of neuropathic pain; these effects were also contingent on PPAR-alpha expression and were observed following either acute or subchronic PPAR-alpha agonist administration. Finally, acute administration of GW7647 and PEA reduced hyperalgesic responses in the complete Freund's adjuvant and carrageenan models of inflammatory pain. Our results suggest that PPAR-alpha agonists may represent a novel class of analgesics.


Assuntos
Analgésicos/farmacologia , PPAR alfa/agonistas , Animais , DNA Complementar/biossíntese , DNA Complementar/genética , Tolerância a Medicamentos , Fenofibrato/análogos & derivados , Fenofibrato/farmacologia , Formaldeído , Hiperalgesia/tratamento farmacológico , Hipolipemiantes/farmacologia , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/efeitos dos fármacos , PPAR alfa/genética , Medição da Dor/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/fisiologia , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Ciática/tratamento farmacológico
9.
J Biol Chem ; 281(32): 22815-8, 2006 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16785227

RESUMO

Oleoylethanolamide (OEA) is an endogenous lipid mediator that inhibits feeding and stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activating receptor-alpha. Little is known about the physiological regulation of this compound outside of the gastrointestinal tract, where its production is regulated by feeding. Here we show that cold exposure increases OEA levels in rat white adipose tissue but not in liver or intestine. This change is accompanied by parallel elevations in the activity of N-acyltransferase, a key enzyme responsible for OEA synthesis, without concomitant changes in fatty acid amide hydrolase, an enzyme responsible for OEA degradation. Moreover, cold stimulates the production of two species of N-oleoylphosphatidylethanolamine OEA precursors. The changes in OEA biosynthesis are reversed by pretreatment with the beta-receptor antagonist propranolol, suggesting a role for beta-adrenoreceptors in this response. In agreement with these findings, the beta-agonists noradrenaline and isoproterenol stimulate OEA production in isolated adipocytes, an effect that is mimicked by the adenylyl cyclase activator forskolin. Collectively, these results identify cold exposure as a natural stimulus for OEA formation in white fat and suggest a role for the sympathetic nervous system in regulating OEA biosynthesis.


Assuntos
Tecido Adiposo/metabolismo , Ácidos Oleicos/biossíntese , Aciltransferases/metabolismo , Adipócitos/metabolismo , Animais , Temperatura Baixa , Colforsina/farmacologia , Endocanabinoides , Epididimo/metabolismo , Isoproterenol/metabolismo , Metabolismo dos Lipídeos , Masculino , Norepinefrina/farmacologia , Ratos , Ratos Wistar , Receptores Adrenérgicos beta/metabolismo
10.
Life Sci ; 77(14): 1685-98, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15963531

RESUMO

Palmitoylethanolamide (PEA), the naturally occurring amide of ethanolamine and palmitic acid, is an endogenous lipid that modulates pain and inflammation. Although the anti-inflammatory effects of PEA were first characterized nearly 50 years ago, the identity of the receptor mediating these actions has long remained elusive. We recently identified the ligand-activated transcription factor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha), as the receptor mediating the anti-inflammatory actions of this lipid amide. Here we outline the history of PEA, starting with its initial discovery in the 1950s, and discuss the pharmacological properties of this compound, particularly in regards to its ability to activate PPAR-alpha.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , PPAR alfa/metabolismo , Ácidos Palmíticos/metabolismo , Ácidos Palmíticos/farmacologia , Receptores de Canabinoides/isolamento & purificação , Receptores de Canabinoides/metabolismo , Amidas , Analgésicos/metabolismo , Anti-Inflamatórios não Esteroides/história , Endocanabinoides , Etanolaminas , História do Século XX , História do Século XXI , Modelos Biológicos , Ácidos Palmíticos/química , Farmacologia/história
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...