Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Neurobiol Dis ; 58: 132-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23726845

RESUMEN

Neuroplastic changes in the dorsal striatum participate in the transition from casual to habitual drug use and might play a critical role in the development of methamphetamine (METH) addiction. We examined the influence of METH self-administration on gene and protein expression that may form substrates for METH-induced neuronal plasticity in the dorsal striatum. Male Sprague-Dawley rats self-administered METH (0.1mg/kg/injection, i.v.) or received yoked saline infusions during eight 15-h sessions and were euthanized 2h, 24h, or 1month after cessation of METH exposure. Changes in gene and protein expression were assessed using microarray analysis, RT-PCR and Western blots. Chromatin immunoprecipitation (ChIP) followed by PCR was used to examine epigenetic regulation of METH-induced transcription. METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. METH also caused changes in ΔFosB, BDNF and TrkB protein levels, with increases after 2 and 24h, but decreases after 1month of drug abstinence. Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. These findings show that METH-induced changes in gene expression are mediated, in part, by pCREB-dependent epigenetic phenomena. Thus, METH self-administration might trigger epigenetic changes that mediate alterations in expression of genes and proteins serving as substrates for addiction-related synaptic plasticity.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Estimulantes del Sistema Nervioso Central/administración & dosificación , Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Metanfetamina/administración & dosificación , Trastornos Relacionados con Sustancias/patología , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Estimulantes del Sistema Nervioso Central/efectos adversos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Dopamina/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Ácido Hidroxiindolacético/metabolismo , Masculino , Metanfetamina/efectos adversos , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Dopaminérgicos/metabolismo , Autoadministración , Serotonina/metabolismo , Trastornos Relacionados con Sustancias/etiología , Trastornos Relacionados con Sustancias/fisiopatología , Factores de Tiempo
2.
Int J Neuropsychopharmacol ; 15(9): 1265-74, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21939589

RESUMEN

Effects of varenicline (Champix), a nicotinic partial agonist, were evaluated on subjective effects of nicotine (drug discrimination), motivation for nicotine taking (progressive-ratio schedule of intravenous nicotine self-administration) and reinstatement (cue-induced reinstatement of previously extinguished nicotine-seeking behaviour). Effects on motor performance were assessed in rats trained to discriminate nicotine (0.4 mg/kg) from saline under a fixed-ratio (FR 10) schedule of food delivery and in rats trained to respond for food under a progressive-ratio schedule. At short pretreatment times (5-40 min), varenicline produced full or high levels of partial generalization to nicotine's discriminative-stimulus effects and disrupted responding for food, while there were low levels of partial generalization and no disruption of responding for food at 2- or 4-h pretreatment times. Varenicline (1 and 3 mg/kg, 2-h pretreatment time) enhanced discrimination of low doses of nicotine and to a small extent decreased discrimination of the training dose of nicotine. It also dose-dependently decreased nicotine-taking behaviour, but had no effect on food-taking behaviour under progressive-ratio schedules. Finally, varenicline significantly reduced the ability of a nicotine-associated cue to reinstate extinguished nicotine-seeking behaviour. The ability of varenicline to reduce both nicotine-taking and nicotine-seeking behaviour can contribute to its relatively high efficacy in treating human smokers.


Asunto(s)
Benzazepinas/farmacología , Señales (Psicología) , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Quinoxalinas/farmacología , Tabaquismo/tratamiento farmacológico , Animales , Condicionamiento Operante/efectos de los fármacos , Interpretación Estadística de Datos , Discriminación en Psicología/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Conducta Alimentaria/efectos de los fármacos , Masculino , Motivación/efectos de los fármacos , Nicotina/farmacología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Esquema de Refuerzo , Autoadministración , Tabaquismo/psicología , Vareniclina
3.
J Pharmacol Exp Ther ; 332(3): 1054-63, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19940104

RESUMEN

sigma-1 Receptors are endoplasmic reticulum (ER) chaperones that are implicated in the neuroplasticity associated with psychostimulant abuse. We immunocytochemically examined the distribution of sigma-1 receptors in the brain of drug-naive rats and then examined the dynamics of sigma-1 receptors and other ER chaperones in specific brain subregions of rats that self-administered methamphetamine, received methamphetamine passively, or received only saline injections. sigma-1 Receptors were found to be expressed in moderate to high levels in the olfactory bulb, striatum, nucleus accumbens shell, olfactory tubercle, amygdala, hippocampus, red nucleus, ventral tegmental area, substantia nigra, and locus ceruleus. Methamphetamine, whether self-administered or passively received, significantly elevated ER chaperones including the sigma-1 receptor, BiP, and calreticulin in the ventral tegmental area and substantia nigra. In the olfactory bulb, however, only the sigma-1 receptor chaperone was increased, and this increase occurred only in rats that actively self-administered methamphetamine. Consistent with an increase in sigma-1 receptors, extracellular signal-regulated kinase was found to be activated and protein kinase A attenuated in the olfactory bulb of methamphetamine self-administering rats. sigma-1 Receptors in the olfactory bulb were found to be colocalized with dopamine D1 receptors. These results indicate that methamphetamine induces ER stress in the ventral tegmental area and substantia nigra in rats whether the drug is received actively or passively. However, the changes seen only in rats that actively self-administered methamphetamine suggest that D1 and sigma-1 receptors in the olfactory bulb might play an important role in the motivational conditioning/learning aspects of methamphetamine self-administration in the rat.


Asunto(s)
Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Retículo Endoplásmico/metabolismo , Metanfetamina/farmacología , Chaperonas Moleculares/biosíntesis , Receptores sigma/biosíntesis , Animales , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Células CHO , Estimulantes del Sistema Nervioso Central/administración & dosificación , Cricetinae , Cricetulus , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Masculino , Metanfetamina/administración & dosificación , Ratas , Ratas Sprague-Dawley , Autoadministración , Receptor Sigma-1
4.
J Pharmacol Exp Ther ; 327(2): 482-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18725543

RESUMEN

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.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Ácidos Araquidónicos/metabolismo , Benzamidas/farmacología , Carbamatos/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Dopamina/análisis , Nicotina/farmacología , Núcleo Accumbens/efectos de los fármacos , Alcamidas Poliinsaturadas/metabolismo , Tabaquismo/tratamiento farmacológico , Amidohidrolasas/fisiología , Animales , Endocannabinoides , Hidrólisis , Masculino , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/química , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Recompensa , Autoadministración , Tabaquismo/enzimología
5.
Neuropharmacology ; 55(2): 190-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18547596

RESUMEN

The striatum contains a high density of histamine H(3) receptors, but their role in striatal function is poorly understood. Previous studies have demonstrated antagonistic interactions between striatal H(3) and dopamine D(1) receptors at the biochemical level, while contradictory results have been reported about interactions between striatal H(3) and dopamine D(2) receptors. In this study, by using reserpinized mice, we demonstrate the existence of behaviorally significant antagonistic postsynaptic interactions between H(3) and D(1) and also between H(3) and dopamine D(2) receptors. The selective H(3) receptor agonist imetit inhibited, while the H(3) receptor antagonist thioperamide potentiated locomotor activation induced by either the D(1) receptor agonist SKF 38393 or the D(2) receptor agonist quinpirole. High scores of locomotor activity were obtained with H(3) receptor blockade plus D(1) and D(2) receptor co-activation, i.e., when thioperamide was co-administered with both SKF 38393 and quinpirole. Radioligand binding experiments in striatal membrane preparations showed the existence of a strong and selective H(3)-D(2) receptor interaction at the membrane level. In agonist/antagonist competition experiments, stimulation of H(3) receptors with several H(3) receptor agonists significantly decreased the affinity of D(2) receptors for the agonist. This kind of intramembrane receptor-receptor interactions are a common biochemical property of receptor heteromers. In fact, by using Bioluminescence Resonance Energy Transfer techniques in co-transfected HEK-293 cells, H(3) (but not H(4)) receptors were found to form heteromers with D(2) receptors. This study demonstrates an important role of postsynaptic H(3) receptors in the modulation of dopaminergic transmission by means of a negative modulation of D(2) receptor function.


Asunto(s)
Cuerpo Estriado/fisiología , Receptores de Dopamina D2/fisiología , Receptores Histamínicos H3/fisiología , Inhibidores de Captación Adrenérgica/farmacología , Animales , Conducta Animal/efectos de los fármacos , Línea Celular Transformada , Membrana Celular/efectos de los fármacos , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , AMP Cíclico/metabolismo , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Transferencia de Energía/fisiología , Histamínicos/farmacología , Humanos , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ensayo de Unión Radioligante/métodos , Reserpina/farmacología , Transfección
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.
Neuropsychopharmacology ; 38(7): 1198-208, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23314220

RESUMEN

Although it is more common for drug abuse to progress from tobacco to cannabis, in many cases cannabis use develops before tobacco use. Epidemiological evidence indicates that prior cannabis use increases the likelihood of becoming dependent on tobacco. To determine whether this effect might be due to cannabis exposure per se, in addition to any genetic, social, or environmental factors that might contribute, we extended our series of studies on 'gateway drug' effects in animal models of drug abuse. Rats were exposed to THC, the main psychoactive constituent of cannabis, for 3 days (two intraperitoneal injections/day). Then, starting 1 week later, they were allowed to self-administer nicotine intravenously. THC exposure increased the likelihood of acquiring the nicotine self-administration response from 65% in vehicle-exposed rats to 94% in THC-exposed rats. When the price of nicotine was manipulated by increasing the response requirement, THC-exposed rats maintained higher levels of intake than vehicle-exposed rats, indicating that THC exposure increased the value of nicotine reward. These results contrast sharply with our earlier findings that prior THC exposure did not increase the likelihood of rats acquiring either heroin or cocaine self-administration, nor did it increase the reward value of these drugs. The findings obtained here suggest that a history of cannabis exposure might have lasting effects that increase the risk of becoming addicted to nicotine.


Asunto(s)
Conducta Adictiva/inducido químicamente , Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Nicotina/farmacología , Animales , Conducta Adictiva/psicología , Condicionamiento Operante/efectos de los fármacos , Sinergismo Farmacológico , Masculino , Actividad Motora/efectos de los fármacos , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/farmacología , Ratas , Esquema de Refuerzo , Recompensa , Autoadministración
8.
Nat Neurosci ; 16(11): 1652-61, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24121737

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Ácido Quinurénico/metabolismo , Trastornos Relacionados con Sustancias , Analgésicos/administración & dosificación , Animales , Benzoxazinas/administración & dosificación , Agonistas de Receptores de Cannabinoides/farmacología , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Discriminación en Psicología/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Morfolinas/administración & dosificación , Naftalenos/administración & dosificación , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Refuerzo en Psicología , Saimiri , Prevención Secundaria , Autoadministración , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/patología , Sulfonamidas/farmacología , Tiazoles/farmacología , Factores de Tiempo , Vigilia
9.
Neuropsychopharmacology ; 37(3): 685-96, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22030716

RESUMEN

Since cloning of the dopamine receptor D4 (DRD4), its role in the brain has remained unclear. It has been reported that polymorphism of the DRD4 gene in humans is associated with reactivity to cues related to tobacco smoking. However, the role of DRD4 in animal models of nicotine addiction has seldom been explored. In our study, male Long-Evans rats learned to intravenously self-administer nicotine under a fixed-ratio (FR) schedule of reinforcement. Effects of the selective DRD4 antagonist L-745,870 were evaluated on nicotine self-administration behavior and on reinstatement of extinguished nicotine-seeking behavior induced by nicotine-associated cues or by priming injections of nicotine. L-745,870 was also tested on reinstatement of extinguished food-seeking behavior as a control. In addition, the selective DRD4 agonist PD 168,077 was tested for its ability to reinstate extinguished nicotine-seeking behavior. Finally, L-745,870 was tested in Sprague Dawley rats trained to discriminate administration of 0.4 mg/kg nicotine from vehicle under an FR schedule of food delivery. L-745,870 significantly attenuated reinstatement of nicotine-seeking induced by both nicotine-associated cues and nicotine priming. In contrast, L-745,870 did not affect established nicotine self-administration behavior or reinstatement of food-seeking behavior induced by food cues or food priming. L-745,870 did not produce nicotine-like discriminative-stimulus effects and did not alter discriminative-stimulus effects of nicotine. PD 168,077 did not reinstate extinguished nicotine-seeking behavior. As DRD4 blockade by L-745,870 selectively attenuated both cue- and nicotine-induced reinstatement of nicotine-seeking behavior, without affecting cue- or food-induced reinstatement of food-seeking behavior, DRD4 antagonists are potential therapeutic agents against tobacco smoking relapse.


Asunto(s)
Conducta Animal/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Nicotina/administración & dosificación , Receptores de Dopamina D4/antagonistas & inhibidores , Animales , Aprendizaje Discriminativo/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Masculino , Piridinas/farmacología , Pirroles/farmacología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración
10.
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
11.
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
12.
PLoS One ; 5(1): e8790, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20098750

RESUMEN

Methamphetamine (meth) is an illicit psychostimulant that is abused throughout the world. Repeated passive injections of the drug given in a single day or over a few days cause significant and long-term depletion of dopamine and serotonin in the mammalian brain. Because meth self-administration may better mimic some aspects of human drug-taking behaviors, we examined to what extent this pattern of drug treatment might also result in damage to monoaminergic systems in the brain. Rats were allowed to intravenously self-administer meth (yoked control rats received vehicle) 15 hours per day for 8 days before being euthanized at either 24 hours or at 7 and 14 days after cessation of drug taking. Meth self-administration by the rats was associated with a progressive escalation of daily drug intake to 14 mg/kg per day. Animals that self-administered meth exhibited dose-dependent decreases in striatal dopamine levels during the period of observation. In addition, there were significant reductions in the levels of striatal dopamine transporter and tyrosine hydroxylase proteins. There were also significant decreases in the levels of dopamine, dopamine transporter, and tyrosine hydroxylase in the cortex. In contrast, meth self-administration caused only transient decreases in norepinephrine and serotonin levels in the two brain regions, with these values returning to normal at seven days after cessation of drug taking. Importantly, meth self-administration was associated with significant dose-dependent increases in glial fibrillary acidic protein in both striatum and cortex, with these changes being of greater magnitude in the striatum. These results suggest that meth self-administration by rats is associated with long-term biochemical changes that are reminiscent of those observed in post-mortem brain tissues of chronic meth abusers.


Asunto(s)
Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Metanfetamina/administración & dosificación , Animales , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Metanfetamina/farmacología , Ratas , Autoadministración , Serotonina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
13.
Psychopharmacology (Berl) ; 203(2): 355-67, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18688601

RESUMEN

RATIONALE: Adenosine receptors are involved in cocaine and methamphetamine discrimination and exposure to caffeine can affect behavioral effects of nicotine in rats. OBJECTIVES: Here we investigated the relative involvement of adenosine A(1) and A(2A) receptors in nicotine, cocaine, and methamphetamine discrimination, before and/or during chronic caffeine exposure. MATERIALS AND METHODS: The nonselective adenosine receptor antagonist caffeine, the A(1)-receptor antagonist cyclopentyltheophylline (CPT), and the A(2A)-receptor antagonist MSX-3 were evaluated in rats trained to discriminate 0.4 mg/kg nicotine from saline under a fixed-ratio schedule of food delivery. Effects of adenosine receptor antagonists were then compared in rats discriminating nicotine, methamphetamine, or cocaine from saline during chronic caffeine exposure in their drinking water. RESULTS: Caffeine, CPT, and MSX-3 partially generalized to nicotine and shifted nicotine dose-response curves leftwards. During chronic caffeine exposure, however, all three ligands failed to generalize to nicotine and failed to shift nicotine dose-response curves. In previous experiments, CPT and MSX-3 partially generalized to methamphetamine and cocaine and shifted dose-response curves leftwards. In the present experiments, CPT neither generalized nor shifted dose-response curves for methamphetamine or cocaine during chronic caffeine exposure. However, MSX-3 partially generalized to both psychostimulants and shifted their dose-response curves leftwards. Caffeine partially generalized to cocaine, but not methamphetamine, and shifted both dose-response curves leftwards. CONCLUSIONS: Both adenosine A(1) and A(2A) receptors are capable of modulating the discriminative-stimulus effects of nicotine. Chronic caffeine exposure produces complete tolerance to both A(1)- and A(2A)-mediated effects in nicotine-trained rats. In contrast, chronic caffeine exposure produces tolerance to adenosine A(1)-mediated, but not A(2A)-mediated, effects in methamphetamine- and cocaine-trained rats.


Asunto(s)
Cafeína/farmacología , Cocaína/farmacología , Discriminación en Psicología/efectos de los fármacos , Metanfetamina/farmacología , Nicotina/farmacología , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Agonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A1 , Agonistas del Receptor de Adenosina A2 , Antagonistas del Receptor de Adenosina A2 , Animales , Cafeína/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
Behav Pharmacol ; 19(1): 13-20, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18195590

RESUMEN

The effects of topiramate, a potential treatment for drug dependence, were evaluated in two groups of rats trained to discriminate the administration of either 0.4 mg/kg nicotine or 10 mg/kg cocaine from that of saline, under a fixed-ratio 10 schedule of food delivery. Topiramate (1-60 mg/kg, intraperitoneal) did not produce any nicotine-like or cocaine-like discriminative effects by itself and did not produce any shift in the dose-response curves for nicotine or cocaine discrimination. Thus, the ability to discriminate the effects of nicotine or cocaine does not appear to be altered by topiramate administration. Furthermore, topiramate, given either alone or in combination with nicotine or cocaine, did not depress rates of responding. These experiments indicate that topiramate does not enhance or reduce the ability of rats to discriminate the effects of nicotine or cocaine.


Asunto(s)
Anticonvulsivantes/farmacología , Cocaína/farmacología , Discriminación en Psicología/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Fructosa/análogos & derivados , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Fructosa/administración & dosificación , Fructosa/farmacología , Inyecciones Intraperitoneales , Masculino , Ratas , Ratas Sprague-Dawley , Topiramato
15.
J Biol Chem ; 283(38): 26016-25, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18644790

RESUMEN

The function of dopamine D(3) receptors present in the striatum has remained elusive. In the present study evidence is provided for the existence of dopamine D(1)-D(3) receptor heteromers and for an intramembrane D(1)-D(3) receptor cross-talk in living cells and in the striatum. The formation of D(1)-D(3) receptor heteromers was demonstrated by fluorescence resonance energy transfer and bioluminescence resonance energy transfer techniques in transfected mammalian cells. In membrane preparations from these cells, a synergistic D(1)-D(3) intramembrane receptor-receptor interaction was observed, by which D(3) receptor stimulation enhances D(1) receptor agonist affinity, indicating that the D(1)-D(3) intramembrane receptor-receptor interaction is a biochemical characteristic of the D(1)-D(3) receptor heteromer. The same biochemical characteristic was also observed in membrane preparations from brain striatum, demonstrating the striatal co-localization and heteromerization of D(1) and D(3) receptors. According to the synergistic D(1)-D(3) intramembrane receptor-receptor interaction, experiments in reserpinized mice showed that D(3) receptor stimulation potentiates D(1) receptor-mediated behavioral effects by a different mechanism than D(2) receptor stimulation. The present study shows that a main functional significance of the D(3) receptor is to obtain a stronger dopaminergic response in the striatal neurons that co-express the two receptors.


Asunto(s)
Cuerpo Estriado/metabolismo , Receptores de Dopamina D1/química , Receptores de Dopamina D3/química , Animales , Encéfalo/metabolismo , Membrana Celular/metabolismo , Dimerización , Dopamina/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Neuronas/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA