Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Addict Biol ; 16(3): 405-15, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21054689

ABSTRACT

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.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Cocaine/pharmacology , Dronabinol/pharmacology , Polyunsaturated Alkamides/pharmacology , Psychotropic Drugs/pharmacology , Receptor, Adenosine A2A/drug effects , Receptor, Cannabinoid, CB1/agonists , Xanthines/pharmacology , Animals , Corpus Striatum/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Endocannabinoids , Injections, Intravenous , Male , Marijuana Abuse/physiopathology , Marijuana Abuse/rehabilitation , Microdialysis , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Presynaptic/drug effects , Reinforcement, Psychology , Saimiri , Self Administration
2.
J Pharmacol Exp Ther ; 327(2): 482-90, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18725543

ABSTRACT

Emerging evidence suggests that the rewarding, abuse-related effects of nicotine are modulated by the endocannabinoid system of the brain. For example, pharmacological blockade or genetic deletion of cannabinoid CB(1) receptors can reduce or eliminate many abuse-related behavioral and neurochemical effects of nicotine. Furthermore, doses of Delta(9)-tetrahydrocannabinol and nicotine that are ineffective when given alone can induce conditioned place preference when given together. These previous studies have used systemically administered CB(1) receptor agonists and antagonists and gene deletion techniques, which affect cannabinoid CB(1) receptors throughout the brain. A more functionally selective way to alter endocannabinoid activity is to inhibit fatty acid amide hydrolase (FAAH), thereby magnifying and prolonging the effects of the endocannabinoid anandamide only when and where it is synthesized and released on demand. Here, we combined behavioral and neurochemical approaches to evaluate whether the FAAH inhibitor URB597 (cyclohexyl carbamic acid 3'-carbamoyl-3-yl ester) could alter the abuse-related effects of nicotine in rats. We found that URB597, at a dose (0.3 mg/kg) that had no behavioral effects by itself, prevented development of nicotine-induced conditioned place preference (CPP) and acquisition of nicotine self-administration. URB597 also reduced nicotine-induced reinstatement in both CPP and self-administration models of relapse. Furthermore, in vivo microdialysis showed that URB597 reduced nicotine-induced dopamine elevations in the nucleus accumbens shell, the terminal area of the brain's mesolimbic reward system. These findings suggest that FAAH inhibition can counteract the addictive properties of nicotine and that FAAH may serve as a new target for development of medications for treatment of tobacco dependence.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arachidonic Acids/metabolism , Benzamides/pharmacology , Carbamates/pharmacology , Conditioning, Psychological/drug effects , Dopamine/analysis , Nicotine/pharmacology , Nucleus Accumbens/drug effects , Polyunsaturated Alkamides/metabolism , Tobacco Use Disorder/drug therapy , Amidohydrolases/physiology , Animals , Endocannabinoids , Hydrolysis , Male , Motor Activity/drug effects , Nucleus Accumbens/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reward , Self Administration , Tobacco Use Disorder/enzymology
3.
J Neurosci ; 27(21): 5615-20, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17522306

ABSTRACT

Increasing use of cannabis makes the search for medications to reduce cannabis abuse extremely important. Here, we show that homomeric alpha7 nicotinic receptors are novel molecular entities that could be targeted in the development of new drugs for the treatment of cannabis dependence. In rats, systemic administration of the selective alpha7 nicotinic acetylcholine receptor antagonist methyllycaconitine (MLA), but not the selective heteromeric non-alpha7 nicotinic acetylcholine receptor antagonist dihydrobetaerythroidine, (1) antagonized the discriminative effects of delta-9-tetrahydrocannabinol (THC), the main active ingredient in cannabis, (2) reduced intravenous self-administration of the synthetic cannabinoid CB1 receptor agonist WIN55,212-2 [(R)-(+)-[2,3-dihydro-5-methyl-3[(4-morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone, mesylate salt], and (3) decreased THC-induced dopamine elevations in the shell of the nucleus accumbens. Altogether, our results indicate that blockade of alpha7 nicotinic receptors reverses abuse-related behavioral and neurochemical effects of cannabinoids. Importantly, MLA reversed the effects of cannabinoids at doses that did not produce depressant or toxic effects, further pointing to alpha7 nicotinic antagonists as potentially useful agents in the treatment of cannabis abuse in humans.


Subject(s)
Drug Delivery Systems/methods , Marijuana Abuse/drug therapy , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Benzoxazines/administration & dosage , Discrimination Learning/drug effects , Discrimination Learning/physiology , Male , Marijuana Abuse/physiopathology , Morpholines/administration & dosage , Motor Activity/drug effects , Motor Activity/physiology , Naphthalenes/administration & dosage , Nicotinic Antagonists/therapeutic use , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Nicotinic/physiology , Self Administration , alpha7 Nicotinic Acetylcholine Receptor
4.
Neuropsychopharmacology ; 32(11): 2249-59, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17356572

ABSTRACT

The mechanism of action responsible for the motor depressant effects of cannabinoids, which operate through centrally expressed cannabinoid CB1 receptors, is still a matter of debate. In the present study, we report that CB1 and adenosine A2A receptors form heteromeric complexes in co-transfected HEK-293T cells and rat striatum, where they colocalize in fibrilar structures. In a human neuroblastoma cell line, CB1 receptor signaling was found to be completely dependent on A2A receptor activation. Accordingly, blockade of A2A receptors counteracted the motor depressant effects produced by the intrastriatal administration of a cannabinoid CB1 receptor agonist. These biochemical and behavioral findings demonstrate that the profound motor effects of cannabinoids depend on physical and functional interactions between striatal A2A and CB1 receptors.


Subject(s)
Cannabinoids/pharmacology , Corpus Striatum/drug effects , Motor Activity/drug effects , Receptor, Adenosine A2A/physiology , Receptor, Cannabinoid, CB1/physiology , Adenosine A2 Receptor Agonists , Analysis of Variance , Animals , Arachidonic Acids/pharmacology , Behavior, Animal , Cannabinoids/agonists , Cannabinoids/antagonists & inhibitors , Cell Line, Transformed , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Drug Interactions , Humans , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/drug effects , Rats , Rats, Wistar , Receptor, Adenosine A2A/deficiency , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/deficiency , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...