Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 42(8): 1619-1629, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28139681

RESUMO

The currently available antismoking medications have limited efficacy and often fail to prevent relapse. Thus, there is a pressing need for newer, more effective treatment strategies. Recently, we demonstrated that enhancing endogenous levels of kynurenic acid (KYNA, a neuroinhibitory product of tryptophan metabolism) counteracts the rewarding effects of cannabinoids by acting as a negative allosteric modulator of α7 nicotinic receptors (α7nAChRs). As the effects of KYNA on cannabinoid reward involve nicotinic receptors, in the present study we used rat and squirrel monkey models of reward and relapse to examine the possibility that enhancing KYNA can counteract the effects of nicotine. To assess specificity, we also examined models of cocaine reward and relapse in monkeys. KYNA levels were enhanced by administering the kynurenine 3-monooxygenase (KMO) inhibitor, Ro 61-8048. Treatment with Ro 61-8048 decreased nicotine self-administration in rats and monkeys, but did not affect cocaine self-administration. In rats, Ro 61-8048 reduced the ability of nicotine to induce dopamine release in the nucleus accumbens shell, a brain area believed to underlie nicotine reward. Perhaps most importantly, Ro 61-8048 prevented relapse-like behavior when abstinent rats or monkeys were reexposed to nicotine and/or cues that had previously been associated with nicotine. Ro 61-8048 was also effective in monkey models of cocaine relapse. All of these effects of Ro 61-8048 in monkeys, but not in rats, were reversed by pretreatment with a positive allosteric modulator of α7nAChRs. These findings suggest that KMO inhibition may be a promising new approach for the treatment of nicotine addiction.


Assuntos
Ácido Cinurênico/metabolismo , Nicotina/farmacologia , Reforço Psicológico , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Animais , Cocaína/administração & dosagem , Cocaína/farmacologia , Dopamina/metabolismo , Isoxazóis/farmacologia , Masculino , Nicotina/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Ratos , Recidiva , Saimiri , Prevenção Secundária , Autoadministração , Sulfonamidas/antagonistas & inibidores , Tiazóis/antagonistas & inibidores
2.
Neuropsychopharmacology ; 41(9): 2283-93, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26888056

RESUMO

Nicotine, the main psychoactive component of tobacco, and (-)-Δ(9)-tetrahydrocannabinol (THC), the main psychoactive ingredient in cannabis, play major roles in tobacco and marijuana dependence as reinforcers of drug-seeking and drug-taking behavior. Drugs that act as inverse agonists of cannabinoid CB1 receptors in the brain can attenuate the rewarding and abuse-related effects of nicotine and THC, but their clinical use is hindered by potentially serious side effects. The recently developed CB1-receptor neutral antagonists may provide an alternative therapeutic approach to nicotine and cannabinoid dependence. Here we compare attenuation of nicotine and THC reinforcement and reinstatement in squirrel monkeys by the CB1-receptor inverse agonist rimonabant and by the recently developed CB1-receptor neutral antagonist AM4113. Both rimonabant and AM4113 reduced two effects of nicotine and THC that play major roles in tobacco and marijuana dependence: (1) maintenance of high rates of drug-taking behavior, and (2) priming- or cue-induced reinstatement of drug-seeking behavior in abstinent subjects (models of relapse). In contrast, neither rimonabant nor AM4113 modified cocaine-reinforced or food-reinforced operant behavior under similar experimental conditions. However, both rimonabant and AM4113 reduced cue-induced reinstatement in monkeys trained to self-administer cocaine, suggesting the involvement of a common cannabinoid-mediated mechanism in the cue-induced reinstatement for different drugs of abuse. These findings point to CB1-receptor neutral antagonists as a new class of medications for treatment of both tobacco dependence and cannabis dependence.


Assuntos
Dronabinol/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Nicotina/administração & dosagem , Piperidinas/administração & dosagem , Pirazóis/administração & dosagem , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Reforço Psicológico , Animais , Condicionamento Operante/efeitos dos fármacos , Sinais (Psicologia) , Masculino , Abuso de Maconha/prevenção & controle , Recidiva , Rimonabanto , Saimiri , Autoadministração , Tabagismo/prevenção & controle
3.
Psychopharmacology (Berl) ; 233(10): 1867-77, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26803499

RESUMO

RATIONALE: N-(4-hydroxyphenyl)-arachidonamide (AM404) is an anandamide transport inhibitor shown to reduce rewarding and relapse-inducing effects of nicotine in several animal models of tobacco dependence. However, the reinforcing/rewarding effects of AM404 are not clear. OBJECTIVES: We investigated whether AM404 maintains self-administration behavior or reinstates extinguished drug seeking in squirrel monkeys. METHODS AND RESULTS: In monkeys with a history of anandamide or cocaine self-administration, we substituted injections of AM404 (1-100 µg/kg/injection). Using a 10-response, fixed-ratio schedule, self-administration behavior was maintained by AM404. Dose-response curves had inverted U shapes, with peak response rates occurring at a dose of 10 µg/kg/injection. In anandamide-experienced monkeys, we also demonstrated self-administration of another anandamide transport inhibitor VDM11. In addition to supporting self-administration, priming injections of AM404 (0.03-0.3 mg/kg) reinstated drug-seeking behavior previously reinforced by cannabinoids (∆(9)-tetrahydrocannabinol (THC) or anandamide) or cocaine. Both AM404 self-administration behavior and reinstatement of drug seeking by AM404 were reduced by treatment with the cannabinoid CB1 receptor antagonist/inverse agonist rimonabant (0.3 mg/kg). Moreover, the reinforcing effects of AM404 were potentiated by the treatment with the fatty acid amide hydrolase (FAAH) inhibitor URB597 (0.3 mg/kg) suggesting a major role of anandamide in these effects. Finally, AM404 (0.3 mg/kg) potentiated the reinforcing effects of anandamide but not those of cocaine. CONCLUSIONS: In non-human primates, AM404 effectively reinforced self-administration behavior and induced reinstatement of drug-seeking behavior in abstinent monkeys. These effects appeared to be mediated by cannabinoid CB1 receptors. Therefore, compounds that promote actions of endocannabinoids throughout the brain by inhibiting their membrane transport may have a potential for abuse.


Assuntos
Ácidos Araquidônicos/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Animais , Ácidos Araquidônicos/antagonistas & inibidores , Ácidos Araquidônicos/metabolismo , Benzamidas/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Carbamatos/farmacologia , Cocaína/farmacologia , Relação Dose-Resposta a Droga , Dronabinol/farmacologia , Endocanabinoides/antagonistas & inibidores , Endocanabinoides/metabolismo , Masculino , Nicotina/farmacologia , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/antagonistas & inibidores , Alcamidas Poli-Insaturadas/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Pirazóis/farmacologia , Reforço Psicológico , Recompensa , Rimonabanto , Saimiri , Autoadministração
4.
Psychopharmacology (Berl) ; 233(10): 1791-800, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26149611

RESUMO

RATIONALE: Group II metabotropic glutamate receptors (mGluR2 and mGluR3) have been suggested to play an important role in mediation of drug-reinforced behaviors, as well as in the mechanisms underlying relapse in abstinent subjects. The prototypical mGluR2/3 agonist, LY379268, has been shown to attenuate nicotine reinforcement and cue-induced reinstatement of drug seeking in rats, as well as reinstatement induced by drug-associated stimuli and contexts across different drugs of abuse (i.e., cocaine, heroin, and methamphetamine). However, in primates, LY379268 has been shown to produce conflicting results on abuse-related effects of cocaine, and there are no data available for nicotine. OBJECTIVES: To explore the therapeutic potential of mGluR2/3 agonists, we compared the effects of LY379268 (0.03-1.0 mg/kg) on nicotine, cocaine, and food self-administration under a fixed-ratio (FR10) schedule in three separate groups of squirrel monkeys. Moreover, we studied the effects of LY379268 on nicotine/cocaine priming-induced and cue-induced reinstatement of drug-seeking behavior in nicotine- and cocaine-experienced groups of animals. RESULTS: LY379268 blocked nicotine, but not cocaine, self-administration in monkeys. There was a partial overlap between doses that affected nicotine and food self-administration. In abstinent monkeys, LY379268 dose-dependently blocked nicotine, but not cocaine, priming-induced reinstatement of drug seeking. In both cocaine-experienced and nicotine-experienced groups of animals, LY379268 potently reduced cue-induced reinstatement of drug-seeking behavior. CONCLUSIONS: The present findings provide strong support for the potential utility of mGlu2/3 receptor agonists for the treatment of nicotine dependence and suggest their utility for prevention of relapse induced by environmental cues associated with drug taking.


Assuntos
Aminoácidos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Cocaína/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Nicotina/administração & dosagem , Tabagismo/tratamento farmacológico , Animais , Sinais (Psicologia) , Relação Dose-Resposta a Droga , Interações Medicamentosas , Masculino , Receptores de Glutamato Metabotrópico/agonistas , Recidiva , Reforço Psicológico , Saimiri , Autoadministração
5.
Biol Psychiatry ; 78(7): 452-62, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25802079

RESUMO

BACKGROUND: Based on rodent studies, group II metabotropic glutamate receptors (mGluR2 and mGluR3) were suggested as targets for addiction treatment. However, LY379268 and other group II agonists do not discriminate between the mainly presynaptic inhibitory mGluR2 (the proposed treatment target) and mGluR3. These agonists also produce tolerance over repeated administration and are no longer considered for addiction treatment. Here, we determined the effects of AZD8529, a selective positive allosteric modulator of mGluR2, on abuse-related effects of nicotine in squirrel monkeys and rats. METHODS: We first assessed modulation of mGluR2 function by AZD8529 using functional in vitro assays in membranes prepared from a cell line expressing human mGluR2 and in primate brain slices. We then determined AZD8529 (.03-10 mg/kg, intramuscular injection) effects on intravenous nicotine self-administration and reinstatement of nicotine seeking induced by nicotine priming or nicotine-associated cues. We also determined AZD8529 effects on food self-administration in monkeys and nicotine-induced dopamine release in accumbens shell in rats. RESULTS: AZD8529 potentiated agonist-induced activation of mGluR2 in the membrane-binding assay and in primate cortex, hippocampus, and striatum. In monkeys, AZD8529 decreased nicotine self-administration at doses (.3-3 mg/kg) that did not affect food self-administration. AZD8529 also reduced nicotine priming- and cue-induced reinstatement of nicotine seeking after extinction of the drug-reinforced responding. In rats, AZD8529 decreased nicotine-induced accumbens dopamine release. CONCLUSIONS: These results provide evidence for efficacy of positive allosteric modulators of mGluR2 in nonhuman primate models of nicotine reinforcement and relapse. This drug class should be considered for nicotine addiction treatment.


Assuntos
Comportamento de Procura de Droga/efeitos dos fármacos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Indóis/farmacologia , Oxidiazóis/farmacologia , Tabagismo/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Cricetulus , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Comportamento de Procura de Droga/fisiologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/sangue , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Células HEK293 , Humanos , Indóis/sangue , Masculino , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Oxidiazóis/sangue , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Saimiri , Autoadministração , Tabagismo/fisiopatologia
6.
Neuropsychopharmacology ; 40(9): 2185-97, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25754762

RESUMO

Inhibition of the enzyme fatty acid amide hydrolase (FAAH) counteracts reward-related effects of nicotine in rats, but it has not been tested for this purpose in non-human primates. Therefore, we studied the effects of the first- and second-generation O-arylcarbamate-based FAAH inhibitors, URB597 (cyclohexyl carbamic acid 3'-carbamoyl-3-yl ester) and URB694 (6-hydroxy-[1,1'-biphenyl]-3-yl-cyclohexylcarbamate), in squirrel monkeys. Both FAAH inhibitors: (1) blocked FAAH activity in brain and liver, increasing levels of endogenous ligands for cannabinoid and α-type peroxisome proliferator-activated (PPAR-α) receptors; (2) shifted nicotine self-administration dose-response functions in a manner consistent with reduced nicotine reward; (3) blocked reinstatement of nicotine seeking induced by reexposure to either nicotine priming or nicotine-associated cues; and (4) had no effect on cocaine or food self-administration. The effects of FAAH inhibition on nicotine self-administration and nicotine priming-induced reinstatement were reversed by the PPAR-α antagonist, MK886. Unlike URB597, which was not self-administered by monkeys in an earlier study, URB694 was self-administered at a moderate rate. URB694 self-administration was blocked by pretreatment with an antagonist for either PPAR-α (MK886) or cannabinoid CB1 receptors (rimonabant). In additional experiments in rats, URB694 was devoid of THC-like or nicotine-like interoceptive effects under drug-discrimination procedures, and neither of the FAAH inhibitors induced dopamine release in the nucleus accumbens shell--consistent with their lack of robust reinforcing effects in monkeys. Overall, both URB597 and URB694 show promise for the initialization and maintenance of smoking cessation because of their ability to block the rewarding effects of nicotine and prevent nicotine priming-induced and cue-induced reinstatement.


Assuntos
Benzamidas/farmacologia , Carbamatos/farmacologia , Oxigenases de Função Mista/antagonistas & inibidores , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Recompensa , Animais , Compostos de Bifenilo/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Sinais (Psicologia) , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Comportamento de Procura de Droga/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Masculino , Oxigenases de Função Mista/metabolismo , Modelos Animais , Ratos , Ratos Sprague-Dawley , Recidiva , Saimiri , Autoadministração , Fatores de Tempo
7.
J Neurosci ; 34(19): 6480-4, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24806674

RESUMO

Different doses of an adenosine A2A receptor antagonist MSX-3 [3,7-dihydro-8-[(1E)-2-(3-ethoxyphenyl)ethenyl]-7 methyl-3-[3-(phosphooxy)propyl-1-(2 propynil)-1H-purine-2,6-dione] were found previously to either decrease or increase self-administration of cannabinoids delta-9-tetrahydrocannabinol (THC) or anandamide in squirrel monkeys. It was hypothesized that the decrease observed with a relatively low dose of MSX-3 was related to blockade of striatal presynaptic A2A receptors that modulate glutamatergic neurotransmission, whereas the increase observed with a higher dose was related to blockade of postsynaptic A2A receptors localized in striatopallidal neurons. This hypothesis was confirmed in the present study by testing the effects of the preferential presynaptic and postsynaptic A2A receptor antagonists SCH-442416 [2-(2-furanyl)-7-[3-(4-methoxyphenyl)propyl]-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine] and KW-6002 [(E)-1, 3-diethyl-8-(3,4-dimethoxystyryl)-7-methyl-3,7-dihydro-1H-purine-2,6-dione], respectively, in squirrel monkeys trained to intravenously self-administer THC. SCH-442416 produced a significant shift to the right of the THC self-administration dose-response curves, consistent with antagonism of the reinforcing effects of THC. Conversely, KW-6002 produced a significant shift to the left, consistent with potentiation of the reinforcing effects of THC. These results show that selectively blocking presynaptic A2A receptors could provide a new pharmacological approach to the treatment of marijuana dependence and underscore corticostriatal glutamatergic neurotransmission as a possible main mechanism involved in the rewarding effects of THC.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Dronabinol/farmacologia , Receptor A2A de Adenosina/efeitos dos fármacos , Receptores Pré-Sinápticos/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Dronabinol/antagonistas & inibidores , Masculino , Abuso de Maconha/tratamento farmacológico , Purinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Reforço Psicológico , Recompensa , Saimiri , Autoadministração , Xantinas/farmacologia
8.
Nat Neurosci ; 16(11): 1652-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24121737

RESUMO

In the reward circuitry of the brain, α-7-nicotinic acetylcholine receptors (α7nAChRs) modulate effects of Δ(9)-tetrahydrocannabinol (THC), marijuana's main psychoactive ingredient. Kynurenic acid (KYNA) is an endogenous negative allosteric modulator of α7nAChRs. Here we report that the kynurenine 3-monooxygenase (KMO) inhibitor Ro 61-8048 increases brain KYNA levels and attenuates cannabinoid-induced increases in extracellular dopamine in reward-related brain areas. In the self-administration model of drug abuse, Ro 61-8048 reduced the rewarding effects of THC and the synthetic cannabinoid WIN 55,212-2 in squirrel monkeys and rats, respectively, and it also prevented relapse to drug-seeking induced by reexposure to cannabinoids or cannabinoid-associated cues. The effects of enhancing endogenous KYNA levels with Ro 61-8048 were prevented by positive allosteric modulators of α7nAChRs. Despite a clear need, there are no medications approved for treatment of marijuana dependence. Modulation of KYNA offers a pharmacological strategy for achieving abstinence from marijuana and preventing relapse.


Assuntos
Encéfalo/metabolismo , Ácido Cinurênico/metabolismo , Transtornos Relacionados ao Uso de Substâncias , Analgésicos/administração & dosagem , Animais , Benzoxazinas/administração & dosagem , Agonistas de Receptores de Canabinoides/farmacologia , Condicionamento Operante/efeitos dos fármacos , Sinais (Psicologia) , Discriminação Psicológica/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Dronabinol/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Morfolinas/administração & dosagem , Naftalenos/administração & dosagem , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Reforço Psicológico , Saimiri , Prevenção Secundária , Autoadministração , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/patologia , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Fatores de Tempo , Vigília
9.
Addict Biol ; 18(1): 30-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22264200

RESUMO

Although substantial research effort has focused on developing pharmacological treatments for cocaine abuse, no effective medications have been developed. Recent studies show that enzymes that metabolize cocaine in the periphery, forestalling its entry into the brain, can prevent cocaine toxicity and its behavioral effects in rodents. Here we report on effects of one such enzyme (Albu-CocH) on the pharmacokinetic and behavioral effects of cocaine in squirrel monkeys. Albu-CocH was developed from successive mutations of human butyrylcholinesterase (BChE) and has 1000-fold greater catalytic activity against cocaine than naturally occurring BChE. Pharmacokinetic studies showed that Albu-CocH (5 mg/kg) had a half-life of 56.6 hours in squirrel monkeys. In these studies, plasma levels of cocaine following i.v. 1 mg/kg cocaine were reduced 2 hours after administration of Albu-CocH, whereas plasma levels of the cocaine metabolite ecgonine methyl ester were increased. These effects were still evident 72 hours following Albu-CocH administration. In behavioral experiments in monkeys, pre-treatment with 5 mg/kg Albu-CocH dramatically decreased self-administration of a reinforcing dose of i.v. cocaine (30 µg/kg/injection) for over 24 hours. Pre-treatment with 5 mg/kg Albu-CocH also attenuated the reinstatement of extinguished cocaine self-administration by an i.v. priming injection of cocaine (0.1 or 0.3 mg/kg) and, in separate studies, attenuated the discriminative-stimulus effects of cocaine. The ability of Albu-CocH to attenuate the abuse-related effects of cocaine in squirrel monkeys indicates that further investigation of BChE mutants as potential treatment for cocaine abuse and toxicity is warranted.


Assuntos
Albuminas/farmacologia , Butirilcolinesterase/farmacologia , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Cocaína/farmacocinética , Inibidores da Captação de Dopamina/farmacocinética , Albuminas/farmacocinética , Análise de Variância , Animais , Formação de Anticorpos/efeitos dos fármacos , Biocatálise , Butirilcolinesterase/farmacocinética , Cocaína/administração & dosagem , Cocaína/antagonistas & inibidores , Aprendizagem por Discriminação/efeitos dos fármacos , Inibidores da Captação de Dopamina/administração & dosagem , Inibidores da Captação de Dopamina/antagonistas & inibidores , Relação Dose-Resposta a Droga , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Comportamento de Procura de Droga/efeitos dos fármacos , Meia-Vida , Humanos , Masculino , Reforço Psicológico , Saimiri , Autoadministração
10.
Neuropsychopharmacology ; 37(8): 1838-47, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22453137

RESUMO

Experimental drugs that activate α-type peroxisome proliferator-activated receptors (PPARα) have recently been shown to reduce the rewarding effects of nicotine in animals, but these drugs have not been approved for human use. The fibrates are a class of PPARα-activating medications that are widely prescribed to improve lipid profiles and prevent cardiovascular disease, but these drugs have not been tested in animal models of nicotine reward. Here, we examine the effects of clofibrate, a representative of the fibrate class, on reward-related behavioral, electrophysiological, and neurochemical effects of nicotine in rats and squirrel monkeys. Clofibrate prevented the acquisition of nicotine-taking behavior in naive animals, substantially decreased nicotine taking in experienced animals, and counteracted the relapse-inducing effects of re-exposure to nicotine or nicotine-associated cues after a period of abstinence. In the central nervous system, clofibrate blocked nicotine's effects on neuronal firing in the ventral tegmental area and on dopamine release in the nucleus accumbens shell. All of these results suggest that fibrate medications might promote smoking cessation. The fact that fibrates are already approved for human use could expedite clinical trials and subsequent implementation of fibrates as a treatment for tobacco dependence, especially in smokers with abnormal lipid profiles.


Assuntos
Clofibrato/farmacologia , Avaliação Pré-Clínica de Medicamentos/psicologia , Hipolipemiantes/farmacologia , Nicotina/farmacologia , Recompensa , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Clofibrato/uso terapêutico , Modelos Animais de Doenças , Dopamina/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Indóis/farmacologia , Masculino , Neurônios/fisiologia , Nicotina/administração & dosagem , Nicotina/antagonistas & inibidores , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , PPAR alfa/agonistas , PPAR alfa/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Saimiri , Prevenção Secundária , Autoadministração , Tabagismo/tratamento farmacológico , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
11.
J Neurosci ; 31(19): 7043-8, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21562266

RESUMO

Two endogenous ligands for cannabinoid CB1 receptors, anandamide (N-arachidonoylethanolamine) and 2-arachidonoylglycerol (2-AG), have been identified and characterized. 2-AG is the most prevalent endogenous cannabinoid ligand in the brain, and electrophysiological studies suggest 2-AG, rather than anandamide, is the true natural ligand for cannabinoid receptors and the key endocannabinoid involved in retrograde signaling in the brain. Here, we evaluated intravenously administered 2-AG for reinforcing effects in nonhuman primates. Squirrel monkeys that previously self-administered anandamide or nicotine under a fixed-ratio schedule with a 60 s timeout after each injection had their self-administration behavior extinguished by vehicle substitution and were then given the opportunity to self-administer 2-AG. Intravenous 2-AG was a very effective reinforcer of drug-taking behavior, maintaining higher numbers of self-administered injections per session and higher rates of responding than vehicle across a wide range of doses. To assess involvement of CB1 receptors in the reinforcing effects of 2-AG, we pretreated monkeys with the cannabinoid CB(1) receptor inverse agonist/antagonist rimonabant [N-piperidino-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methylpyrazole-3-carboxamide]. Rimonabant produced persistent blockade of 2-AG self-administration without affecting responding maintained by food under similar conditions. Thus, 2-AG was actively self-administered by monkeys with or without a history of cannabinoid self-administration, and the reinforcing effects of 2-AG were mediated by CB1 receptors. Self-administration of 2-AG by squirrel monkeys provides a valuable procedure for studying abuse liability of medications that interfere with 2-AG signaling within the brain and for investigating mechanisms involved in the reinforcing effects of endocannabinoids.


Assuntos
Ácidos Araquidônicos/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Glicerídeos/administração & dosagem , Motivação/efeitos dos fármacos , Análise de Variância , Animais , Endocanabinoides , Extinção Psicológica/efeitos dos fármacos , Infusões Intravenosas , Injeções Intravenosas , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Reforço Psicológico , Rimonabanto , Saimiri , Autoadministração
12.
Biol Psychiatry ; 69(7): 633-41, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20801430

RESUMO

BACKGROUND: Recent findings indicate that inhibitors of fatty acid amide hydrolase (FAAH) counteract the rewarding effects of nicotine in rats. Inhibition of FAAH increases levels of several endogenous substances in the brain, including the endocannabinoid anandamide and the noncannabinoid fatty acid ethanolamides oleoylethanolamide (OEA) and palmitoylethanolamide, which are ligands for alpha-type peroxisome proliferator-activated nuclear receptors (PPAR-α). Here, we evaluated whether directly acting PPAR-α agonists can modulate reward-related effects of nicotine. METHODS: We combined behavioral, neurochemical, and electrophysiological approaches to evaluate effects of the PPAR-α agonists [[4-Chloro-6-[(2,3-dimethylphenyl)amino]-2-pyrimidinyl]thio]acetic acid (WY14643) and methyl oleoylethanolamide (methOEA; a long-lasting form of OEA) on 1) nicotine self-administration in rats and squirrel monkeys; 2) reinstatement of nicotine-seeking behavior in rats and monkeys; 3) nicotine discrimination in rats; 4) nicotine-induced electrophysiological activity of ventral tegmental area dopamine neurons in anesthetized rats; and 5) nicotine-induced elevation of dopamine levels in the nucleus accumbens shell of freely moving rats. RESULTS: The PPAR-α agonists dose-dependently decreased nicotine self-administration and nicotine-induced reinstatement in rats and monkeys but did not alter food- or cocaine-reinforced operant behavior or the interoceptive effects of nicotine. The PPAR-α agonists also dose-dependently decreased nicotine-induced excitation of dopamine neurons in the ventral tegmental area and nicotine-induced elevations of dopamine levels in the nucleus accumbens shell of rats. The ability of WY14643 and methOEA to counteract the behavioral, electrophysiological, and neurochemical effects of nicotine was reversed by the PPAR-α antagonist 1-[(4-Chlorophenyl)methyl]-3-[(1,1-dimethylethyl)thio]-a,a-dimethyl-5-(1-methylethyl)-1H-Indole-2-propanoic acid (MK886). CONCLUSIONS: These findings indicate that PPAR-α might provide a valuable new target for antismoking medications.


Assuntos
Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , PPAR alfa/metabolismo , Reforço Psicológico , Recompensa , Potenciais de Ação/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Masculino , Microdiálise/métodos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Oligossacarídeos/farmacologia , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Saimiri , Autoadministração , Área Tegmentar Ventral/citologia
13.
Addict Biol ; 16(3): 405-15, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21054689

RESUMO

Several recent studies suggest functional and molecular interactions between striatal adenosine A(2A) and cannabinoid CB(1) receptors. Here, we demonstrate that A(2A) receptors selectively modulate reinforcing effects of cannabinoids. We studied effects of A(2A) receptor blockade on the reinforcing effects of delta-9-tetrahydrocannabinol (THC) and the endogenous CB(1) receptor ligand anandamide under a fixed-ratio schedule of intravenous drug injection in squirrel monkeys. A low dose of the selective adenosine A(2A) receptor antagonist MSX-3 (1 mg/kg) caused downward shifts of THC and anandamide dose-response curves. In contrast, a higher dose of MSX-3 (3 mg/kg) shifted THC and anandamide dose-response curves to the left. MSX-3 did not modify cocaine or food pellet self-administration. Also, MSX-3 neither promoted reinstatement of extinguished drug-seeking behavior nor altered reinstatement of drug-seeking behavior by non-contingent priming injections of THC. Finally, using in vivo microdialysis in freely-moving rats, a behaviorally active dose of MSX-3 significantly counteracted THC-induced, but not cocaine-induced, increases in extracellular dopamine levels in the nucleus accumbens shell. The significant and selective results obtained with the lower dose of MSX-3 suggest that adenosine A(2A) antagonists acting preferentially at presynaptic A(2A) receptors might selectively reduce reinforcing effects of cannabinoids that lead to their abuse. However, the appearance of potentiating rather than suppressing effects on cannabinoid reinforcement at the higher dose of MSX-3 would likely preclude the use of such a compound as a medication for cannabis abuse. Adenosine A(2A) antagonists with more selectivity for presynaptic versus postsynaptic receptors could be potential medications for treatment of cannabis abuse.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Ácidos Araquidônicos/farmacologia , Moduladores de Receptores de Canabinoides/farmacologia , Cocaína/farmacologia , Dronabinol/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Psicotrópicos/farmacologia , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor CB1 de Canabinoide/agonistas , Xantinas/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Endocanabinoides , Injeções Intravenosas , Masculino , Abuso de Maconha/fisiopatologia , Abuso de Maconha/reabilitação , Microdiálise , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Pré-Sinápticos/efeitos dos fármacos , Reforço Psicológico , Saimiri , Autoadministração
14.
Biol Psychiatry ; 64(11): 930-7, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18814866

RESUMO

BACKGROUND: CB(1) cannabinoid receptors in the brain are known to participate in the regulation of reward-based behaviors. However, the contribution of each of the endocannabinoid transmitters, anandamide and 2-arachidonoylglycerol (2-AG), to these behaviors remains undefined. To address this question, we assessed the effects of URB597, a selective anandamide deactivation inhibitor, as a reinforcer of drug-seeking and drug-taking behavior in squirrel monkeys. METHODS: We investigated the reinforcing effects of the fatty acid amide hydrolase (FAAH) inhibitor URB597 in monkeys trained to intravenously self-administer Delta(9)-tetrahydrocannabinol (THC), anandamide, or cocaine and quantified brain endocannabinoid levels using liquid chromatography/mass spectrometry. We measured brain FAAH activity using an ex vivo enzyme assay. RESULTS: URB597 (.3 mg/kg, intravenous) blocked FAAH activity and increased anandamide levels throughout the monkey brain. This effect was accompanied by a marked compensatory decrease in 2-AG levels. Monkeys did not self-administer URB597, and the drug did not promote reinstatement of extinguished drug-seeking behavior previously maintained by THC, anandamide, or cocaine. Pretreatment with URB597 did not modify self-administration of THC or cocaine, even though, as expected, it significantly potentiated anandamide self-administration. CONCLUSIONS: In the monkey brain, the FAAH inhibitor URB597 increases anandamide levels while causing a compensatory down-regulation in 2-AG levels. These effects are accompanied by a striking lack of reinforcing properties, which distinguishes URB597 from direct-acting cannabinoid agonists such as THC. Our results reveal an unexpected functional heterogeneity within the endocannabinoid signaling system and suggest that FAAH inhibitors might be used therapeutically without risk of abuse or triggering of relapse to drug abuse.


Assuntos
Amidoidrolases/metabolismo , Ácidos Araquidônicos/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Dronabinol/administração & dosagem , Alcamidas Poli-Insaturadas/administração & dosagem , Reforço Psicológico , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Benzamidas/farmacologia , Encéfalo/anatomia & histologia , Moduladores de Receptores de Canabinoides/metabolismo , Carbamatos/farmacologia , Cromatografia Líquida/métodos , Cocaína/administração & dosagem , Relação Dose-Resposta a Droga , Endocanabinoides , Masculino , Ratos , Ratos Wistar , Esquema de Reforço , Saimiri , Autoadministração/métodos
15.
Neuropsychopharmacology ; 33(12): 2870-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18305459

RESUMO

Accumulating evidence suggests the endocannabinoid system modulates environmental cues' ability to induce seeking of drugs, including nicotine and alcohol. However, little attention has been directed toward extending these advances to the growing problem of cannabis use disorders. Therefore, we studied intravenous self-administration of Delta(9)-tetrahydrocannabinol (THC), the main psychoactive constituent of marijuana, using a second-order schedule of drug seeking. Squirrel monkeys' lever responses produced only a brief cue light until the end of the session, when the final response delivered THC along with the cue. When a reinstatement procedure was used to model relapse following a period of abstinence, THC-seeking behavior was robustly reinstated by the cue or by pre-session administration of THC, other cannabinoid agonists, or morphine, but not cocaine. The cannabinoid antagonist rimonabant blocked cue-induced drug seeking, THC-induced drug seeking, and the direct reinforcing effects of THC. Thus, rimonabant and related medications might be effective as treatments for cannabinoid dependence.


Assuntos
Química Encefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Dronabinol/antagonistas & inibidores , Abuso de Maconha/tratamento farmacológico , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Química Encefálica/fisiologia , Moduladores de Receptores de Canabinoides/agonistas , Moduladores de Receptores de Canabinoides/antagonistas & inibidores , Moduladores de Receptores de Canabinoides/metabolismo , Sinais (Psicologia) , Modelos Animais de Doenças , Esquema de Medicação , Masculino , Abuso de Maconha/metabolismo , Abuso de Maconha/fisiopatologia , Morfina/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Reforço Psicológico , Rimonabanto , Saimiri , Prevenção Secundária , Autoadministração
16.
Psychopharmacology (Berl) ; 183(4): 413-21, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16292593

RESUMO

L-Deprenyl (selegiline) is used in the treatment of Parkinson's disease and has been proposed as an aid for cigarette smoking cessation and a treatment for psychostimulant abuse. L-Deprenyl is metabolized in the body to L-methamphetamine and L-amphetamine, suggesting that it may have abuse potential. The current study assessed whether L-deprenyl or its isomer would maintain drug-seeking behavior on a second-order schedule and whether L-deprenyl would alter drug-seeking behavior maintained by D-amphetamine if given as a pretreatment. Squirrel monkeys learned to respond on a second-order schedule of reinforcement, where every tenth response was followed by a brief light flash, and the first brief light flash after 30 min was paired with intravenous (i.v.) injection of D-amphetamine (0.56 mg/kg), administered over a 2-min period at the end of the session. When responding was stable, saline or different i.v. doses of D-amphetamine (0.3-1.0 mg/kg), L-deprenyl (0.1-10.0 mg/kg), and D-deprenyl (0.1-3.0 mg/kg) were substituted for 10 days each. Subsequently, monkeys were pretreated with 0.3 or 1.0 mg/kg L-deprenyl intramuscularly 30 min prior to D-amphetamine baseline sessions. D-Amphetamine maintained high rates of drug-seeking behavior on the second-order schedule. D-Deprenyl maintained high rates of drug-seeking behavior similar to D-amphetamine. L-Deprenyl maintained lower rates of responding that were not significantly above saline substitution levels. Pretreatment with L-deprenyl failed to alter drug-seeking behavior maintained by D-amphetamine. These results indicate that D-deprenyl, but not L-deprenyl, may have abuse potential. Under conditions where drug-seeking and drug-taking behaviors are actively maintained by D-amphetamine, L-deprenyl, at doses that specifically inhibit type B monoamine oxidase, may not be effective as a treatment.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Operante/efeitos dos fármacos , Dextroanfetamina/farmacologia , Inibidores da Monoaminoxidase/farmacologia , Selegilina/farmacologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Animais , Relação Dose-Resposta a Droga , Masculino , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/química , Esquema de Reforço , Saimiri , Selegilina/química , Autoadministração , Estereoisomerismo
17.
J Neurosci ; 25(23): 5645-50, 2005 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-15944392

RESUMO

Anandamide, an endogenous ligand for brain cannabinoid CB(1) receptors, produces many behavioral effects similar to those of Delta(9)-tetrahydrocannabinol (THC), the main psychoactive ingredient in marijuana. Reinforcing effects of THC have been demonstrated in experimental animals, but there is only indirect evidence that endogenous cannabinoids such as anandamide participate in brain reward processes. We now show that anandamide serves as an effective reinforcer of drug-taking behavior when self-administered intravenously by squirrel monkeys. We also show that methanandamide, a synthetic long-lasting anandamide analog, similarly serves as a reinforcer of drug-taking behavior. Finally, we show that the reinforcing effects of both anandamide and methanandamide are blocked by pretreatment with the cannabinoid CB(1) receptor antagonist rimonabant (SR141716). These findings strongly suggest that release of endogenous cannabinoids is involved in brain reward processes and that activation of cannabinoid CB(1) receptors by anandamide could be part of the signaling of natural rewarding events.


Assuntos
Ácidos Araquidônicos/farmacologia , Moduladores de Receptores de Canabinoides/farmacologia , Reforço Psicológico , Animais , Ácidos Araquidônicos/administração & dosagem , Ácidos Araquidônicos/química , Moduladores de Receptores de Canabinoides/administração & dosagem , Moduladores de Receptores de Canabinoides/química , Cocaína/administração & dosagem , Cocaína/farmacologia , Endocanabinoides , Infusões Intravenosas , Masculino , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/fisiologia , Rimonabanto , Saimiri , Autoadministração , Estereoisomerismo , Transtornos Relacionados ao Uso de Substâncias/psicologia
18.
Psychopharmacology (Berl) ; 169(2): 135-40, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12827345

RESUMO

RATIONALE: Interest in therapeutic activities of cannabinoids has been restrained by the fact that they are most often mediated through activation of cannabinoid CB1 receptors, the same receptors that mediate the effects of delta9-tetrahydrocannabinol (THC) and are responsible for the abuse liability of marijuana. Persistent intravenous self-administration of THC by animals was first demonstrated in squirrel monkeys and shown to be mediated by CB1 receptors, but monkeys in the study had a history of cocaine self-administration, raising the possibility that persistent neurobiological adaptations might subsequently predispose animals to self-administer THC. OBJECTIVES: To demonstrate persistent intravenous self-administration of THC in drug-naive squirrel monkeys. METHODS: Monkeys with no history of exposure to other drugs learned to press a lever for intravenous injections (0.2 ml in 0.2 s) of THC under a 10-response, fixed-ratio schedule with a 60-s time-out after each injection. Acquisition of THC self-administration was rapid and the final schedule was reached in 11-34 sessions. Dose of THC was then varied from 1 to 16 microg/kg per injection with vehicle extinction following each dose of THC. RESULTS: THC maintained significantly higher numbers of self-administered injections per session and higher rates of responding than vehicle at doses of 2, 4 and 8 microg/kg per injection, with maximal rates of responding at 4 microg/kg per injection. Response rates, injections per session and total THC intake per session were two- to three-fold greater in monkeys with no history of exposure to other drugs compared to previous findings in monkeys with a history of cocaine self-administration. CONCLUSIONS: THC can act as an effective reinforcer of drug-taking behavior in monkeys with no history of exposure to other drugs, suggesting that self-administration of THC by monkeys provides a reliable animal model of human marijuana abuse.


Assuntos
Dronabinol/administração & dosagem , Dronabinol/farmacocinética , Autoadministração/métodos , Animais , Condicionamento Operante/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Injeções Intravenosas , Masculino , Abuso de Maconha/etiologia , Abuso de Maconha/prevenção & controle , Esquema de Reforço , Saimiri
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...