Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Neuropharmacology ; 176: 108170, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32479813

RESUMEN

Nicotine, the primary psychoactive component in tobacco, plays a major role in the initiation and maintenance of tobacco dependence and addiction, a leading cause of preventable death worldwide. An essential need thus exists for more effective pharmacotherapies for nicotine-use cessation. Previous reports suggest that pharmacological and genetic blockade of CB1 receptors attenuate nicotine reinforcement and reward; while exogenous agonists enhanced these abuse-related behaviors. In this study, we utilized complementary genetic and pharmacologic approaches to test the hypothesis that increasing the levels of the endocannabinoid 2-arachindonoylglycerol (2-AG), will enhance nicotine reward by stimulating neuronal CB1 receptors. Contrary to our hypothesis, we found that inhibition of monoacylglycerol lipase (MAGL), the primary catabolic enzyme of 2-AG, attenuates nicotine conditioned place preference (CPP) in mice, through a non-CB1 receptor-mediated mechanism. MAGL inhibition did not alter palatable food reward or Lithium Chloride (LiCl) aversion. In support of our findings, repeated MAGL inhibition did not induce a reduction in CB1 brain receptor levels or hinder function. To explore the potential mechanism of action, we investigated if MAGL inhibition affected other fatty acid levels in our CPP paradigm. Indeed, MAGL inhibition caused a concomitant decrease in arachidonic acid (AA) levels in various brain regions of interest, suggesting an AA cascade-dependent mechanism. This idea is supported by dose-dependent attenuation of nicotine preference by the selective COX-2 inhibitors valdecoxib and LM-4131. Collectively, these findings, along with our reported studies on nicotine withdrawal, suggest that inhibition of MAGL represents a promising new target for the development of pharmacotherapies to treat nicotine dependence.


Asunto(s)
Condicionamiento Clásico/efectos de los fármacos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo , Nicotina/administración & dosificación , Recompensa , Tabaquismo/metabolismo , Animales , Ansiolíticos/farmacología , Ácidos Araquidónicos/farmacología , Benzodioxoles/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Condicionamiento Clásico/fisiología , Endocannabinoides/farmacología , Inhibidores Enzimáticos/farmacología , Glicéridos/farmacología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Piperidinas/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Tabaquismo/tratamiento farmacológico , Tabaquismo/psicología
2.
Int J Neuropsychopharmacol ; 16(8): 1867-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23590881

RESUMEN

Anxiety is one of the early symptoms of opioid withdrawal and contributes to continued drug use and relapse. The acoustic startle response (ASR) is a component of anxiety that has been shown to increase during opioid withdrawal in both humans and animals. We investigated the role of corticotropin-releasing factor (CRF) and norepinephrine (NE), two key mediators of the brain stress system, on acute heroin withdrawal-potentiated ASR. Rats injected with heroin (2 mg/kg s.c.) displayed an increased ASR when tested 4 h after heroin treatment. A similar increase in ASR was found in rats 10-20 h into withdrawal from extended access (12 h) to i.v. heroin self-administration, a model that captures several aspects of heroin addiction in humans. Both the α 2 adrenergic receptor agonist clonidine (10 µg/kg s.c.) and CRF1 receptor antagonist N,N-bis(2-methoxyethyl)-3-(4-methoxy-2-methylphenyl)-2,5-dimethyl-pyrazolo[1,5-a] pyrimidin-7-amine (MPZP; 20 mg/kg s.c.) blocked heroin withdrawal-potentiated startle. To investigate the relationship between CRF1 and α 2 adrenergic receptors in the potentiation of the ASR, we tested the effect of MPZP on yohimbine (1.25 mg/kg s.c.)-potentiated startle and clonidine on CRF (2 µg i.c.v.)-potentiated startle. Clonidine blocked CRF-potentiated startle, whereas MPZP partially attenuated but did not reverse yohimbine-potentiated startle, suggesting that CRF may drive NE release to potentiate startle. These results suggest that CRF1 and α 2 receptors play an important role in the heightened anxiety-like behaviour observed during acute withdrawal from heroin, possibly via CRF inducing the release of NE in stress-related brain regions.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Heroína/efectos adversos , Narcóticos/efectos adversos , Receptores Adrenérgicos alfa 2/metabolismo , Reflejo de Sobresalto/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Estimulación Acústica , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Análisis de Varianza , Animales , Clonidina/farmacología , Condicionamiento Operante/efectos de los fármacos , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/farmacología , Modelos Animales de Enfermedad , Masculino , Norepinefrina/metabolismo , Psicoacústica , Pirimidinas/farmacología , Ratas , Ratas Wistar , Reflejo de Sobresalto/efectos de los fármacos , Autoadministración , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Factores de Tiempo , Yohimbina/farmacología
3.
AAPS J ; 11(2): 342-52, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19430909

RESUMEN

Abstinence symptoms in cannabis-dependent individuals are believed to contribute to the maintenance of regular marijuana use. However, there are currently no medications approved by the FDA to treat cannabis-related disorders. The only treatment currently shown consistently to alleviate cannabinoid withdrawal in both animals and humans is substitution therapy using the psychoactive constituent of marijuana, Delta(9)-tetrahydrocannabinol (THC). However, new genetic and pharmacological tools are available to increase endocannabinoid levels by targeting fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), the enzymes responsible for the degradation of the endogenous cannabinoid ligands anandamide and 2-arachidonoylglycerol, respectively. In the present study, we investigated whether increasing endogenous cannabinoids levels, through the use of FAAH (-/-) mice as well as the FAAH inhibitor URB597 or the MAGL inhibitor JZL184, would reduce the intensity of withdrawal signs precipitated by the CB(1) receptor antagonist rimonabant in THC-dependent mice. Strikingly, acute administration of either URB597 or JZL184 significantly attenuated rimonabant-precipitated withdrawal signs in THC-dependent mice. In contrast, FAAH (-/-) mice showed identical withdrawal responses as wild-type mice under a variety of conditions, suggesting that the absence of this enzyme across the development of dependence and during rimonabant challenge does not affect withdrawal responses. Of importance, subchronic administration of URB597 did not lead to cannabinoid dependence and neither URB597 nor JZL184 impaired rotarod motor coordination. These results support the concept of targeting endocannabinoid metabolizing enzymes as a promising treatment for cannabis withdrawal.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Moduladores de Receptores de Cannabinoides/metabolismo , Dronabinol , Endocannabinoides , Inhibidores Enzimáticos/uso terapéutico , Alucinógenos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Benzamidas/farmacología , Benzodioxoles/farmacología , Moduladores de Receptores de Cannabinoides/antagonistas & inhibidores , Carbamatos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Piperidinas/farmacología , Equilibrio Postural/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Pirazoles/farmacología , Rimonabant , Síndrome de Abstinencia a Sustancias/psicología
4.
AAPS J ; 11(1): 39-44, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19184452

RESUMEN

The endogenous cannabinoid N-arachidonoyl ethanolamine (anandamide; AEA) produces most of its pharmacological effects by binding and activating CB(1) and CB(2) cannabinoid receptors within the CNS and periphery. However, the actions of AEA are short lived because of its rapid catabolism by fatty acid amide hydrolase (FAAH). Indeed, FAAH knockout mice as well as animals treated with FAAH inhibitors are severely impaired in their ability to hydrolyze AEA as well as a variety of noncannabinoid lipid signaling molecules and consequently possess greatly elevated levels of these endogenous ligands. In this mini review, we describe recent research that has investigated the functional consequences of inhibiting this enzyme in a wide range of animal models of inflammatory and neuropathic pain states. FAAH-compromised animals reliably display antinociceptive and anti-inflammatory phenotypes with a similar efficacy as direct-acting cannabinoid receptor agonists, such as Delta(9)-tetrahydrocannabinol (THC), the primary psychoactive constituent of Cannabis sativa. Importantly, FAAH blockade does not elicit any apparent psychomimetic effects associated with THC or produce reinforcing effects that are predictive of human drug abuse. The beneficial effects caused by FAAH blockade in these models are predominantly mediated through the activation of CB(1) and/or CB(2) receptors, though noncannabinoid mechanisms of actions can also play contributory or even primary roles. Collectively, the current body of scientific literature suggests that activating the endogenous cannabinoid system by targeting FAAH is a promising strategy to treat pain and inflammation but lacks untoward side effects typically associated with Cannabis sativa.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Analgésicos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Ácidos Araquidónicos/metabolismo , Inflamación/tratamiento farmacológico , Dolor/tratamiento farmacológico , Alcamidas Poliinsaturadas/metabolismo , Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Amidohidrolasas/fisiología , Analgésicos/toxicidad , Animales , Antiinflamatorios no Esteroideos/toxicidad , Modelos Animales de Enfermedad , Dronabinol/farmacología , Dronabinol/toxicidad , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Endocannabinoides , Glicéridos/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/fisiopatología , Ratones , Ratones Noqueados , Dolor/fisiopatología , Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/fisiología , Ratas , Receptores de Cannabinoides/efectos de los fármacos , Receptores de Cannabinoides/metabolismo , Receptores Opioides/efectos de los fármacos , Receptores Opioides/fisiología , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA