Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Neuropsychopharmacology ; 42(8): 1619-1629, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28139681

RESUMEN

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.


Asunto(s)
Ácido Quinurénico/metabolismo , Nicotina/farmacología , Refuerzo en Psicología , Sulfonamidas/farmacología , Tiazoles/farmacología , Animales , Cocaína/administración & dosificación , Cocaína/farmacología , Dopamina/metabolismo , Isoxazoles/farmacología , Masculino , Nicotina/administración & dosificación , Núcleo Accumbens/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Ratas , Recurrencia , Saimiri , Prevención Secundaria , Autoadministración , Sulfonamidas/antagonistas & inhibidores , Tiazoles/antagonistas & inhibidores
2.
Neuropsychopharmacology ; 41(9): 2283-93, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26888056

RESUMEN

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.


Asunto(s)
Dronabinol/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Nicotina/administración & dosificación , Piperidinas/administración & dosificación , Pirazoles/administración & dosificación , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Refuerzo en Psicología , Animales , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Masculino , Abuso de Marihuana/prevención & control , Recurrencia , Rimonabant , Saimiri , Autoadministración , Tabaquismo/prevención & control
3.
Psychopharmacology (Berl) ; 233(10): 1867-77, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26803499

RESUMEN

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.


Asunto(s)
Ácidos Araquidónicos/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Animales , Ácidos Araquidónicos/antagonistas & inhibidores , Ácidos Araquidónicos/metabolismo , Benzamidas/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Carbamatos/farmacología , Cocaína/farmacología , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/metabolismo , Masculino , Nicotina/farmacología , Piperidinas/farmacología , Alcamidas Poliinsaturadas/antagonistas & inhibidores , Alcamidas Poliinsaturadas/metabolismo , Alcamidas Poliinsaturadas/farmacología , Pirazoles/farmacología , Refuerzo en Psicología , Recompensa , Rimonabant , Saimiri , Autoadministración
4.
Psychopharmacology (Berl) ; 233(10): 1791-800, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26149611

RESUMEN

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.


Asunto(s)
Aminoácidos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Cocaína/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Nicotina/administración & dosificación , Tabaquismo/tratamiento farmacológico , Animales , Señales (Psicología) , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Masculino , Receptores de Glutamato Metabotrópico/agonistas , Recurrencia , Refuerzo en Psicología , Saimiri , Autoadministración
5.
Biol Psychiatry ; 78(7): 452-62, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25802079

RESUMEN

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.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Indoles/farmacología , Oxadiazoles/farmacología , Tabaquismo/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/fisiología , Fármacos actuantes sobre Aminoácidos Excitadores/sangre , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Células HEK293 , Humanos , Indoles/sangre , Masculino , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Oxadiazoles/sangre , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Saimiri , Autoadministración , Tabaquismo/fisiopatología
6.
Neuropsychopharmacology ; 40(9): 2185-97, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25754762

RESUMEN

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.


Asunto(s)
Benzamidas/farmacología , Carbamatos/farmacología , Oxigenasas de Función Mixta/antagonistas & inhibidores , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Recompensa , Animales , Compuestos de Bifenilo/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Señales (Psicología) , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Masculino , Oxigenasas de Función Mixta/metabolismo , Modelos Animales , Ratas , Ratas Sprague-Dawley , Recurrencia , Saimiri , Autoadministración , Factores de Tiempo
7.
J Neurosci ; 34(19): 6480-4, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24806674

RESUMEN

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.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Dronabinol/farmacología , Receptor de Adenosina A2A/efectos de los fármacos , Receptores Presinapticos/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Dronabinol/antagonistas & inhibidores , Masculino , Abuso de Marihuana/tratamiento farmacológico , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Refuerzo en Psicología , Recompensa , Saimiri , Autoadministración , Xantinas/farmacología
8.
Neuropsychopharmacology ; 37(8): 1838-47, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22453137

RESUMEN

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.


Asunto(s)
Clofibrato/farmacología , Evaluación Preclínica de Medicamentos/psicología , Hipolipemiantes/farmacología , Nicotina/farmacología , Recompensa , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Clofibrato/uso terapéutico , Modelos Animales de Enfermedad , Dopamina/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Indoles/farmacología , Masculino , Neuronas/fisiología , Nicotina/administración & dosificación , Nicotina/antagonistas & inhibidores , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , PPAR alfa/agonistas , PPAR alfa/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Saimiri , Prevención Secundaria , Autoadministración , Tabaquismo/tratamiento farmacológico , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología
9.
Biol Psychiatry ; 69(7): 633-41, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20801430

RESUMEN

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.


Asunto(s)
Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , PPAR alfa/metabolismo , Refuerzo en Psicología , Recompensa , Potenciales de Acción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Masculino , Microdiálisis/métodos , Neuronas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Oligosacáridos/farmacología , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Saimiri , Autoadministración , Área Tegmental Ventral/citología
10.
Addict Biol ; 16(3): 405-15, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21054689

RESUMEN

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.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Ácidos Araquidónicos/farmacología , Moduladores de Receptores de Cannabinoides/farmacología , Cocaína/farmacología , Dronabinol/farmacología , Alcamidas Poliinsaturadas/farmacología , Psicotrópicos/farmacología , Receptor de Adenosina A2A/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Xantinas/farmacología , Animales , Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Endocannabinoides , Inyecciones Intravenosas , Masculino , Abuso de Marihuana/fisiopatología , Abuso de Marihuana/rehabilitación , Microdiálisis , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Presinapticos/efectos de los fármacos , Refuerzo en Psicología , Saimiri , Autoadministración
11.
Psychopharmacology (Berl) ; 183(4): 413-21, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16292593

RESUMEN

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.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Dextroanfetamina/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Selegilina/farmacología , Trastornos Relacionados con Sustancias/psicología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/química , Esquema de Refuerzo , Saimiri , Selegilina/química , Autoadministración , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA