Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Br J Pharmacol ; 174(19): 3333-3345, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28718892

RESUMEN

BACKGROUND AND PURPOSE: Methoxetamine (MXE) is a novel psychoactive substance that is emerging on the Internet and induces dissociative effects and acute toxicity. Its pharmacological effects have not yet been adequately investigated. EXPERIMENTAL APPROACH: We examined a range of behavioural effects induced by acute administration of MXE (0.5-5 mg·kg-1 ; i.p.) in rats and whether it causes rapid neuroadaptive molecular changes. KEY RESULTS: MXE (0.5-5 mg·kg-1 ) affected motor activity in a dose- and time-dependent manner, inducing hypermotility and hypomotility at low and high doses respectively. At low and intermediate doses (0.5 and 1 mg·kg-1 ), MXE induced anxious and/or obsessive-compulsive traits (marble burying test), did not significantly increase sociability (social interaction test) or induce spatial anxiety (elevated plus maze test). At a high dose (5 mg·kg-1 ), MXE induced transient analgesia (tail-flick and hot-plate test), decreased social interaction time (social interaction test) and reduced immobility time while increasing swimming activity (forced swim test), suggesting an antidepressant effect. Acute MXE administration did not affect self-grooming behaviour at any dose tested. Immunohistochemical analysis showed that behaviourally active doses of MXE (1 and 5 mg·kg-1 ) increased phosphorylation of ribosomal protein S6 in the medial prefrontal cortex and hippocampus. CONCLUSIONS AND IMPLICATIONS: MXE differentially affected motor activity, behaviour and emotional states in rats, depending on the dose tested. As reported for ketamine, phosphorylation of the ribosomal protein S6 was increased in MXE-treated animals, thus providing a 'molecular snapshot' of rapid neuroadaptive molecular changes induced by behaviourally active doses of MXE.


Asunto(s)
Encéfalo/efectos de los fármacos , Ciclohexanonas/farmacología , Ciclohexilaminas/farmacología , Drogas Ilícitas/farmacología , Psicotrópicos/farmacología , Animales , Ansiedad/inducido químicamente , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Emociones/efectos de los fármacos , Calor , Locomoción/efectos de los fármacos , Masculino , Conducta Obsesiva/inducido químicamente , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/metabolismo , Conducta Social
2.
Mol Psychiatry ; 19(8): 930-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24296975

RESUMEN

Smoking is the most important preventable cause of morbidity and mortality worldwide. Recent genome-wide association studies highlighted a human haplotype on chromosome 15 underlying the risk for tobacco dependence and lung cancer. Several polymorphisms in the CHRNA3-CHRNA5-CHRNB4 cluster coding for the nicotinic acetylcholine receptor (nAChR) α3, α5 and ß4 subunits were implicated. In mouse models, we define a key role in the control of sensitivity to nicotine for the α5 subunit in dopaminergic (DAergic) neurons of the ventral tegmental area (VTA). We first investigated the reinforcing effects of nicotine in drug-naive α5(-/-) mice using an acute intravenous nicotine self-administration task and ex vivo and in vivo electrophysiological recordings of nicotine-elicited DA cell activation. We designed lentiviral re-expression vectors to achieve targeted re-expression of wild-type or mutant α5 in the VTA, in general, or in DA neurons exclusively. Our results establish a crucial role for α5*-nAChRs in DAergic neurons. These receptors are key regulators that determine the minimum nicotine dose necessary for DA cell activation and thus nicotine reinforcement. Finally, we demonstrate that a single-nucleotide polymorphism, the non-synonymous α5 variant rs16969968, frequent in many human populations, exhibits a partial loss of function of the protein in vivo. This leads to increased nicotine consumption in the self-administration paradigm. We thus define a critical link between a human predisposition marker, its expression in DA neurons and nicotine intake.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Nicotina/farmacología , Receptores Nicotínicos/genética , Potenciales de Acción/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Noqueados , Nicotina/administración & dosificación , Polimorfismo de Nucleótido Simple , Refuerzo en Psicología , Autoadministración , Área Tegmental Ventral/efectos de los fármacos
3.
Br J Pharmacol ; 169(4): 820-33, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23072421

RESUMEN

BACKGROUND AND PURPOSE: Binge eating disorder (BED) is characterized by excessive food intake during short periods of time. Recent evidence suggests that alterations in the endocannabinoid signalling could be involved in the pathophysiology of BED. In this study, we investigated whether pharmacological manipulation of endocannabinoid transmission may be effective in modulating the aberrant eating behaviour present in a validated rat model of BED. EXPERIMENTAL APPROACH: Binge-type eating was induced in female rats by providing limited access to an optional source of dietary fat (margarine). Rats were divided into three groups, all with ad libitum access to chow and water: control (C), with no access to margarine; low restriction (LR), with 2 h margarine access 7 days a week; high restriction (HR), with 2 h margarine access 3 days a week. KEY RESULTS: Compared with the LR group, the HR group consumed more margarine and this was accompanied by an increase in body weight. The cannabinoid CB1/CB2 receptor agonist Δ9-tetrahydrocannabinol significantly increased margarine intake selectively in LR rats, while the fatty acid amide hydrolase inhibitor URB597 showed no effect. The CB1 receptor inverse agonist/antagonist rimonabant dose-dependently reduced margarine intake in HR rats. Notably, in HR rats, chronic treatment with a low dose of rimonabant induced a selective long-lasting reduction in margarine intake that did not develop tolerance, and a significant and persistent reduction in body weight. CONCLUSIONS AND IMPLICATIONS: Chronic pharmacological blockade of CB1 receptors reduces binge eating behaviour in female rats and may prove effective in treating BED, with an associated significant reduction in body weight.


Asunto(s)
Trastorno por Atracón/tratamiento farmacológico , Antagonistas de Receptores de Cannabinoides/uso terapéutico , Modelos Animales de Enfermedad , Endocannabinoides/antagonistas & inhibidores , Piperidinas/uso terapéutico , Pirazoles/uso terapéutico , Receptor Cannabinoide CB1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Trastorno por Atracón/inducido químicamente , Trastorno por Atracón/metabolismo , Agonistas de Receptores de Cannabinoides/administración & dosificación , Agonistas de Receptores de Cannabinoides/toxicidad , Antagonistas de Receptores de Cannabinoides/administración & dosificación , Relación Dosis-Respuesta a Droga , Dronabinol/administración & dosificación , Dronabinol/toxicidad , Agonismo Inverso de Drogas , Tolerancia a Medicamentos , Endocannabinoides/agonistas , Endocannabinoides/metabolismo , Ingestión de Energía/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Femenino , Margarina/efectos adversos , Piperidinas/administración & dosificación , Pirazoles/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Rimonabant , Pérdida de Peso/efectos de los fármacos
4.
Br J Pharmacol ; 167(8): 1652-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22762735

RESUMEN

BACKGROUND AND PURPOSE: Pharmacological interventions aimed at restoring the endocannabinoid system functionality have been proposed as potential tools in the treatment of schizophrenia. Based on our previous results suggesting a potential antipsychotic-like profile of the CB(1) receptor inverse agonist/antagonist, AM251, here we further investigated the effect of chronic AM251 administration on the alteration of the sensorimotor gating functions and endocannabinoid levels induced by isolation rearing in rats. EXPERIMENTAL APPROACH: Using the post-weaning social isolation rearing model, we studied its influence on sensorimotor gating functions through the PPI paradigm. The presence of alterations in the endocannabinoid levels as well as in dopamine and glutamate receptor densities was explored in specific brain regions following isolation rearing. The effect of chronic AM251 administration on PPI response and the associated biochemical alterations was assessed. KEY RESULTS: The disrupted PPI response in isolation-reared rats was paralleled by significant alterations in 2-AG content and dopamine and glutamate receptor densities in specific brain regions. Chronic AM251 completely restored normal PPI response in isolated rats. This behavioural recovery was paralleled by the normalization of 2-AG levels in all the brain areas analysed. Furthermore, AM251 partially antagonized isolation-induced changes in dopamine and glutamate receptors. CONCLUSIONS AND IMPLICATIONS: These results demonstrate the efficacy of chronic AM251 treatment in the recovery of isolation-induced disruption of PPI. Moreover, AM251 counteracted the imbalances in the endocannabinoid content, specifically 2-AG levels, and partially reversed the alterations in dopamine and glutamate systems associated with the disrupted behaviour. Together, these findings support the potential antipsychotic-like activity of CB(1) receptor blockade. LINKED ARTICLES: This article is part of a themed section on Cannabinoids. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.167.issue-8.


Asunto(s)
Encéfalo/efectos de los fármacos , Antagonistas de Receptores de Cannabinoides/farmacología , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Filtrado Sensorial/efectos de los fármacos , Estimulación Acústica , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/fisiología , Endocannabinoides/fisiología , Masculino , Ratas , Receptor Cannabinoide CB1/fisiología , Aislamiento Social
5.
Br J Pharmacol ; 163(7): 1550-62, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21518339

RESUMEN

BACKGROUND AND PURPOSE: Opioids and cannabinoids interact in drug addiction and relapse. We investigated the effect of the opioid receptor antagonist naloxone and/or the cannabinoid CB(1) receptor antagonist rimonabant on cannabinoid-induced reinstatement of heroin seeking and on cannabinoid substitution in heroin-abstinent rats. EXPERIMENTAL APPROACH Rats were trained to self-administer heroin (30 µg·kg(-1) per infusion) under a fixed-ratio 1 reinforcement schedule. After extinction of self-administration (SA) behaviour, we confirmed the effect of naloxone (0.1-1 mg·kg(-1)) and rimonabant (0.3-3 mg·kg(-1)) on the reinstatement of heroin seeking induced by priming with the CB(1) receptor agonist WIN55,212-2 (WIN, 0.15-0.3 mg·kg(-1)). Then, in a parallel set of heroin-trained rats, we evaluated whether WIN (12.5 µg·kg(-1) per infusion) SA substituted for heroin SA after different periods of extinction. In groups of rats in which substitution occurred, we studied the effect of both antagonists on cannabinoid intake. KEY RESULTS: Cannabinoid-induced reinstatement of heroin seeking was significantly attenuated by naloxone (1 mg·kg(-1)) and rimonabant (3 mg·kg(-1)) and fully blocked by co-administration of sub-threshold doses of the two antagonists. Moreover, contrary to immediate (1 day) or delayed (90 days) drug substitution, rats readily self-administered WIN when access was given after 7, 14 or 21 days of extinction from heroin, and showed a response rate that was positively correlated with the extinction period. In these animals, cannabinoid intake was increased by naloxone (1 mg·kg(-1)) and decreased by rimonabant (3 mg·kg(-1)). CONCLUSIONS AND IMPLICATIONS: Our findings extend previous research on the crosstalk between cannabinoid and opioid receptors in relapse mechanisms, which suggests a differential role in heroin-seeking reinstatement and cannabinoid substitution in heroin-abstinent rats.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Adictiva/inducido químicamente , Cannabinoides/farmacología , Heroína/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Benzoxazinas/farmacología , Sustitución de Medicamentos/métodos , Sinergismo Farmacológico , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Esquema de Refuerzo , Rimonabant , Autoadministración/métodos
6.
Br J Pharmacol ; 160(3): 724-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20590575

RESUMEN

BACKGROUND AND PURPOSE: Animal and human studies have shown that sex and hormones are key factors in modulating addiction. Previously, we have demonstrated that self-administration of the cannabinoid CB(1) receptor agonist WIN55,212-2 (WIN; 12.5 microg.kg(-1) per infusion) is dependent on sex, intact female rats being more sensitive than males to the reinforcing properties of cannabinoids, and on the oestrous cycle, ovariectomized (OVX) females being less responsive than intact females. EXPERIMENTAL APPROACH: This follow-up study investigated whether sex and ovarian function also affect reinstatement of cannabinoid-seeking in rats after exposure to drug or cue priming. KEY RESULTS: After priming with 0.15 or 0.3 mg.kg(-1) WIN, intact female rats exhibited stronger reinstatement than males and OVX females. Responses of intact female rats were higher than those of male and OVX rats even after priming with a drug-associated visual (Light) or auditory (Tone) cue, or a WIN + Light combination. However, latency to the first response did not differ between intact and OVX female rats, and males showed the longest latency to initiate lever-pressing activity. CONCLUSIONS AND IMPLICATIONS: Our study provides compelling evidence for a pivotal role of sex and the oestrous cycle in modulating cannabinoid-seeking, with ovariectomy diminishing drug and cue-induced reinstatement. However, it is possible that sex differences during self-administration training are responsible for sex differences in reinstatement. Finding that not only drug primings but also acute exposure to drug-associated cues can reinstate responding in rats could have significant implications for the development of pharmacological and behavioural treatments of abstinent female and male marijuana smokers.


Asunto(s)
Benzoxazinas/farmacología , Extinción Psicológica/fisiología , Morfolinas/farmacología , Naftalenos/farmacología , Ovariectomía/psicología , Animales , Conducta Adictiva/psicología , Benzoxazinas/administración & dosificación , Condicionamiento Operante/fisiología , Señales (Psicología) , Extinción Psicológica/efectos de los fármacos , Femenino , Masculino , Morfolinas/administración & dosificación , Naftalenos/administración & dosificación , Ratas , Ratas Endogámicas , Receptor Cannabinoide CB1/agonistas , Autoadministración , Caracteres Sexuales , Factores de Tiempo
7.
Curr Drug Targets ; 11(4): 450-61, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20017729

RESUMEN

Cannabinoids and opioids are known to strictly interact in many physiological and pathological functions, including addiction. The endogenous opioid system is significantly influenced by maternal or perinatal cannabinoid exposure, major changes concerning operant behaviour in adult animals. Copious data suggests that adolescence is also a particularly sensitive period of life not only for the initiation of abusing illicit drugs, but also for the effects that these drugs exert on the neural circuitries leading to drug dependence. This paper examines the role played by the age of drug exposure in the susceptibility to discriminative and reinforcing effects of both cannabinoids and opioids. We first revisited evidence of alterations in the density and functionality of mu-opioid and CB1 cannabinoid receptors in reward-related brain regions caused by either maternal, postnatal, adolescent or adult exposure to opioids and cannabinoids. Then, we reviewed behavioural evidence of the long-term consequences of exposure to opioids and cannabinoids during gestation, postnatal period, adolescence or adulthood, focusing mostly on drug discrimination and self-administration studies. Overall, evidence confirms a neurobiological convergence of the cannabinoid and opioid systems that is manifest at both receptor and behavioural levels. Although discrepant results have been reported, some data support the gateway hypothesis that adolescent cannabis exposure contributes to greater opioid intake in adulthood. However, it should be kept into consideration that in humans genetic, environmental, and social factors could influence the direct neurobiological effects of early cannabis exposure to the progression to adult drug abuse.


Asunto(s)
Conducta Adictiva/etiología , Encéfalo/efectos de los fármacos , Discriminación en Psicología/efectos de los fármacos , Abuso de Marihuana/etiología , Trastornos Relacionados con Opioides/etiología , Efectos Tardíos de la Exposición Prenatal , Automedicación , Adolescente , Adulto , Factores de Edad , Animales , Conducta Adictiva/metabolismo , Conducta Adictiva/psicología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Femenino , Edad Gestacional , Humanos , Masculino , Abuso de Marihuana/metabolismo , Abuso de Marihuana/psicología , Péptidos Opioides/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/psicología , Embarazo , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/metabolismo , Recurrencia , Refuerzo en Psicología , Recompensa , Factores de Riesgo , Adulto Joven
8.
J Neurosci ; 28(47): 12318-27, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020025

RESUMEN

The identification of the molecular mechanisms involved in nicotine addiction and its cognitive consequences is a worldwide priority for public health. Novel in vivo paradigms were developed to match this aim. Although the beta2 subunit of the neuronal nicotinic acetylcholine receptor (nAChR) has been shown to play a crucial role in mediating the reinforcement properties of nicotine, little is known about the contribution of the different alpha subunit partners of beta2 (i.e., alpha4 and alpha6), the homo-pentameric alpha7, and the brain areas other than the ventral tegmental area (VTA) involved in nicotine reinforcement. In this study, nicotine (8.7-52.6 microg free base/kg/inf) self-administration was investigated with drug-naive mice deleted (KO) for the beta2, alpha4, alpha6 and alpha7 subunit genes, their wild-type (WT) controls, and KO mice in which the corresponding nAChR subunit was selectively re-expressed using a lentiviral vector (VEC mice). We show that WT mice, beta2-VEC mice with the beta2 subunit re-expressed exclusively in the VTA, alpha4-VEC mice with selective alpha4 re-expression in the VTA, alpha6-VEC mice with selective alpha6 re-expression in the VTA, and alpha7-KO mice promptly self-administer nicotine intravenously, whereas beta2-KO, beta2-VEC in the substantia nigra, alpha4-KO and alpha6-KO mice do not respond to nicotine. We thus define the necessary and sufficient role of alpha4beta2- and alpha6beta2-subunit containing nicotinic receptors (alpha4beta2*- and alpha6beta2*-nAChRs), but not alpha7*-nAChRs, present in cell bodies of the VTA, and their axons, for systemic nicotine reinforcement in drug-naive mice.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Receptores Nicotínicos/fisiología , Área Tegmental Ventral/metabolismo , Análisis de Varianza , Animales , Autorradiografía/métodos , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Bloqueadores de los Canales de Calcio/farmacocinética , Conotoxinas/farmacocinética , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Isótopos de Yodo/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nicotínicos/deficiencia , Autoadministración/métodos , Receptor Nicotínico de Acetilcolina alfa 7
9.
Mol Cell Endocrinol ; 286(1-2 Suppl 1): S97-S107, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18372102

RESUMEN

The endocannabinoid system is implicated in the regulation of a variety of physiological processes, among which conditioning, motivation, habit forming, memory, learning, and cognition play pivotal roles in drug reinforcement and reward. In this article we will give a synopsis of last developments in research on cannabinoid actions on brain reward circuits coming from behavioral, neurochemical and electrophysiological studies. Central cannabinoid-induced effects as measured by animal models of addiction, in vivo cerebral microdialysis, in vitro and in vivo electrophysiological recording techniques, will be reviewed. Brain sites that have been implicated in the mediation of addictive cannabinoid properties include primarily the ventral tegmental area, the nucleus accumbens, and the medial prefrontal cortex, although the amygdala, the substantia nigra, the globus pallidus, and the hippocampus have also been shown to be critical structures mediating motivational and reinforcing effects of cannabinoids. Putative neurobiological mechanisms underlying these effects will be delineated.


Asunto(s)
Cannabinoides/efectos adversos , Abuso de Marihuana/fisiopatología , Sistema Nervioso/efectos de los fármacos , Humanos , Abuso de Marihuana/metabolismo
10.
Br J Pharmacol ; 152(5): 795-804, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17891164

RESUMEN

BACKGROUND AND PURPOSE: We recently demonstrated the existence of strain differences in self-administration of the cannabinoid CB1 receptor agonist WIN55,212-2 (WIN) by Long Evans (LE) and Lister Hooded (LH) but not Sprague-Dawley (SD) male rats. This follow-up study is aimed at verifying whether sex and ovarian hormones might also be critical factors in the initiation, retention and extinction of WIN self-administration. EXPERIMENTAL APPROACH: LE, LH and SD male and female rats, the latter either intact or bilaterally ovariectomized (OVX), were trained to self-administer WIN (12.5 microg kg(-1) per infusion) under a FR1 reinforcement schedule, using lever-pressing. KEY RESULTS: Data showed that contrary to the findings in SD rats, LE and LH rats developed robust cannabinoid intake, with rates of responding for WIN being constantly higher in intact females than in males (+45 and +42% for LE and LH strains, respectively). In comparison with intact females, OVX females of both strains acquired self-administration at lower rates, displaying slower acquisition, lower drug intake (-42 and -52% for LE and LH, respectively) and longer extinction. CONCLUSIONS AND IMPLICATIONS: These findings provide the first evidence of significant sex differences in cannabinoid self-administration, females acquiring stable WIN intake at higher rates and more rapidly than males. Moreover, when compared to intact females, a lower percentage of LE and LH OVX rats acquired and maintained stable drug intake, suggesting that ovarian hormones might represent a critical factor in modulating the reinforcing effect of cannabinoids.


Asunto(s)
Benzoxazinas/farmacología , Cannabinoides/farmacología , Morfolinas/farmacología , Naftalenos/farmacología , Ovario/efectos de los fármacos , Factores de Edad , Analgésicos/administración & dosificación , Analgésicos/farmacología , Análisis de Varianza , Animales , Benzoxazinas/administración & dosificación , Cannabinoides/administración & dosificación , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Extinción Psicológica/efectos de los fármacos , Femenino , Infusiones Intravenosas , Masculino , Morfolinas/administración & dosificación , Naftalenos/administración & dosificación , Pruebas de Función Ovárica , Ovariectomía/métodos , Ovario/fisiología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Retención en Psicología/efectos de los fármacos , Autoadministración , Caracteres Sexuales , Factores Sexuales , Factores de Tiempo
11.
Eur J Neurosci ; 17(8): 1723-6, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12752390

RESUMEN

Because opioid and cannabinoid systems have been reported to interact in the modulation of addictive behaviour, this study was aimed at investigating the ability of cannabinoid agents to reinstate or prevent heroin-seeking behaviour after a prolonged period of extinction. In rats previously trained to self-administer heroin intravenously, non-contingent non-reinforced priming administrations of heroin and cannabinoids were presented after long-term extinction, and lever pressing following injections was observed. Results showed that: (i) intravenous priming infusions of heroin (0.1 and 0.2 mg/kg) lead to reinstatement of drug-seeking behaviour; (ii) intraperitoneal priming injections of the central cannabinoid receptor agonists R-(+)-(2,3-dihydro-5-methyl-3-[(4-morpholinyl)methyl]pyrol[1,2,3-de]-1,4-benzoxazinyl) (1-naphthalenyl)methanonemesylate (WIN 55,212-2, 0.15 and 0.3 mg/kg) and (-)-cis-3-[2-hydroxy-4(1,1-dimethyl-heptyl)phenyl]-trans-4-(3-hydroxypropyl) cyclohexanol (CP 55,940, 0.05 and 0.1 mg/kg), but not delta9-tetrahydrocannabinol (delta9-THC, 0.1-1.0 mg/kg), effectively restored heroin-seeking behaviour; (iii) intraperitoneal priming injection of the central cannabinoid receptor antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichloro-phenyl)4-methyl-1H-pyrazole-3-carboxamide (SR 141716A, 0.3 mg/kg) did not reinstate responding, but (iv) completely prevented heroin-induced reinstatement of drug-seeking behaviour. Moreover, heroin-seeking behaviour was still present for a few days following cannabinoid primings, indicating a long-lasting effect of cannabinoids on responding for heroin. These findings indicate that relapse to heroin after an extended drug-free period is triggered by cannabinoid agonists and that SR 141716A prevents drug-seeking behaviour, suggesting that the use of the cannabinoid antagonist could have some therapeutic benefits in heroin-induced relapse.


Asunto(s)
Extinción Psicológica/fisiología , Dependencia de Heroína/fisiopatología , Heroína/farmacología , Narcóticos/farmacología , Receptores de Droga/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Cannabinoides/farmacología , Condicionamiento Operante/efectos de los fármacos , Heroína/administración & dosificación , Masculino , Narcóticos/administración & dosificación , Ratas , Receptores de Cannabinoides , Receptores de Droga/agonistas , Receptores de Droga/antagonistas & inhibidores , Autoadministración
12.
Neuroscience ; 110(1): 1-6, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11882367

RESUMEN

Astrocytes have been proved to play a critical role in neuromodulation, neuroprotection, pH maintenance, axon guidance control during development, homeostasis preservation and blood brain barrier maintenance in the CNS (Kimmelberg and Norenberg, 1989). Quantitative changes in the expression of glial fibrillary acidic protein (GFAP), a cytoskeletal intermediate filament protein exclusively expressed in astrocytes (Bignami et al, 1972), have been observed after administration of alcohol (Framke, 1995), morphine (Beitner-Johnson et al., 1993), amphetamine and its derivates (Aguirre et al., 1999), cannabinoids (Suarez et al., 2000), nicotine (Janson and Moller, 1993), caffeine (Marret et al., 1993) and prenatal exposure to cocaine (Clarke et al., 1996; Nassogne et al., 1998). However, the general astrocytic response to drugs of abuse is still far from being defined. In the present study we examined the in vivo astroglial response to cocaine in mouse dentate gyrus, the hippocampus being a common target of neurotoxic agents (Walsh and Emerich, 1988) which has a prominent effect on learning and memory processes (Eichenbaum et al., 1992). Quantitative changes in immunoreactivity of GFAP were investigated 24 h after acute and repeated daily administration of intraperitoneal cocaine (20 mg/kg). Drug-induced morphological alterations and spatial distribution of astrocytes were evaluated by means of confocal microscope. The results show that, compared to control animals, GFAP expression is two-fold enhanced after a single cocaine injection, still significantly higher after seven consecutive daily administrations, but not statistically different after prolonged (14 days) drug treatment. Moreover, morphological and morphometric analyses reveal significant modifications in astrocytic numbers, cell size and shape complexity. These data demonstrate that in mouse dentate gyrus, cocaine exposure differently affects the expression of GFAP and induces strong changes in astrocytes proliferation rate and cell morphology. Taken together, our findings provide the first in vivo quantitative and qualitative evaluation of astrocytic response to several regimens of cocaine in adult animals brain.


Asunto(s)
Astrocitos/efectos de los fármacos , División Celular/efectos de los fármacos , Cocaína/toxicidad , Giro Dentado/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Gliosis/inducido químicamente , Regulación hacia Arriba/efectos de los fármacos , Animales , Astrocitos/metabolismo , Astrocitos/patología , Recuento de Células , División Celular/fisiología , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/fisiología , Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Cocaína/patología , Trastornos Relacionados con Cocaína/fisiopatología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Giro Dentado/patología , Giro Dentado/fisiopatología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Gliosis/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Regulación hacia Arriba/fisiología
13.
Psychopharmacology (Berl) ; 156(4): 410-6, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11498718

RESUMEN

RATIONALE: Delta9-tetrahydrocannabinol (Delta9-THC), the main psychoactive ingredient of marijuana, as well as synthetic cannabinoid (CB1) receptor agonists, has led to negative or equivocal results when tested with the intravenous self-administration procedure, the best validated behavioural model for evaluating abuse liability of drugs in experimental animals. We recently reported, however, that the synthetic CB1 receptor agonist WIN 55,212-2 is intravenously self-administered by drug-naive mice and that its self-administration is blocked by the cannabinoid CB1 receptor antagonist SR 141716A. OBJECTIVE: To assess a reliable model of cannabinoid intravenous self-administration in rats. Long Evans male rats were allowed the opportunity to self-administer WIN 55,212-2 at doses ranging from 6.25 to 50 microg/kg per injection, under a fixed-ratio 1 (FR1) schedule of reinforcement and nose-pokes as the operant responses. The effect of either a change in the unit drug dose available or a pretreatment with the specific CB1 receptor antagonist SR 141716A were then investigated (maintenance phase). Finally, the extinction of the self-administration behaviour was evaluated. RESULTS: Response rate depended on the drug dose available, with maximum rates occurring at 12.5 microg/kg per injection. Response rate increased following pretreatment with the specific CB1 receptor antagonist, SR 141716A. Moreover, operant behaviour rapidly extinguished following both the substitution of saline or vehicle for cannabinoid and the disconnection of the drug delivery pumps. CONCLUSION: Rats will intravenously self-administer the synthetic CB1 receptor agonist WIN 55,212-2 under specific experimental conditions, thus allowing further investigation of the neurobiological mechanisms underlying cannabinoid-taking behaviour.


Asunto(s)
Analgésicos/administración & dosificación , Analgésicos/farmacología , Cannabinoides/metabolismo , Morfolinas/administración & dosificación , Morfolinas/farmacología , Naftalenos/administración & dosificación , Naftalenos/farmacología , Receptores de Droga/agonistas , Esquema de Refuerzo , Animales , Benzoxazinas , Relación Dosis-Respuesta a Droga , Inyecciones Intravenosas , Masculino , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Ratas Long-Evans , Receptores de Cannabinoides , Receptores de Droga/antagonistas & inhibidores , Rimonabant , Autoadministración/psicología , Abuso de Sustancias por Vía Intravenosa/psicología
14.
J Neurosci ; 21(14): 5344-50, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11438610

RESUMEN

The present study was designed to explore the relationship between the cannabinoid and opioid receptors in animal models of opioid-induced reinforcement. The acute administration of SR141716A, a selective central cannabinoid CB1 receptor antagonist, blocked heroin self-administration in rats, as well as morphine-induced place preference and morphine self-administration in mice. Morphine-dependent animals injected with SR141716A exhibited a partial opiate-like withdrawal syndrome that had limited consequences on operant responses for food and induced place aversion. These effects were associated with morphine-induced changes in the expression of CB1 receptor mRNA in specific nuclei of the reward circuit, including dorsal caudate putamen, nucleus accumbens, and septum. Additionally, the opioid antagonist naloxone precipitated a mild cannabinoid-like withdrawal syndrome in cannabinoid-dependent rats and blocked cannabinoid self-administration in mice. Neither SR141716A nor naloxone produced any intrinsic effect on these behavioral models. The present results show the existence of a cross-interaction between opioid and cannabinoid systems in behavioral responses related to addiction and open new strategies for the treatment of opiate dependence.


Asunto(s)
Dependencia de Heroína/metabolismo , Dependencia de Morfina/metabolismo , Receptores de Droga/metabolismo , Receptores Opioides/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Cannabinoides/antagonistas & inhibidores , Núcleo Caudado/metabolismo , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Heroína/administración & dosificación , Heroína/antagonistas & inhibidores , Masculino , Ratones , Morfina/administración & dosificación , Morfina/antagonistas & inhibidores , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/metabolismo , Piperidinas/farmacología , Putamen/metabolismo , Pirazoles/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Cannabinoides , Receptores de Droga/antagonistas & inhibidores , Receptores de Droga/genética , Rimonabant , Autoadministración , Tabique del Cerebro/metabolismo
15.
Neuroreport ; 12(10): 2243-6, 2001 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-11447342

RESUMEN

gamma-Hydroxybutyric acid (GHB) is a widely used recreational drug known to exert positive reinforcing effects in animals and humans. The GABA(B) receptor agonist baclofen has been proved to possess antimotivational effect and to inhibit alcohol, cocaine, heroin and nicotine intake. In the present study we evaluated the effect of baclofen on i.v. self-administration of GHB in drug-naive mice under a fixed-ratio (FR-1) schedule of reinforcement and nose-poking-like response as operandum. Results show that baclofen was able to completely prevent GHB seeking behaviour, decreasing the rate of responding to basal values, without showing any reinforcing properties when made contingent on nose-poking response. Our findings demonstrate that baclofen antagonises GHB i.v. self-administration, supporting an important role for the GABA(B) receptor in reward-related mechanisms underlying addictive behaviour.


Asunto(s)
Baclofeno/farmacología , Agonistas del GABA/farmacología , Hidroxibutiratos/administración & dosificación , Refuerzo en Psicología , Autoadministración , Animales , Baclofeno/uso terapéutico , Conducta Adictiva/tratamiento farmacológico , Conducta Adictiva/psicología , Agonistas del GABA/uso terapéutico , Inyecciones Intravenosas , Masculino , Ratones , Receptores de GABA-B/fisiología , Autoadministración/psicología
16.
Toxicol Lett ; 121(3): 199-206, 2001 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-11369474

RESUMEN

Environmentally relevant levels of lead (Pb) have been demonstrated to have a neurotoxic action, especially on children. In this study, Long-Evans rats were continuously exposed to Pb acetate in drinking water from early gestational days (2-6) or from 28 days of age. At the 13th week of age, the functional activity of the nucleus accumbens (NAC) dopaminergic system was studied by means of transversal microdialysis. Neither Pb treatment regimen modified dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) extracellular concentrations, with respect to control rats. However, neuronal depolarisation, induced by perfusion with 60 mM KCl, increased extracellular DA levels to a significantly minor degree in rats exposed to Pb during the intrauterine life, with respect to both control and adult Pb treated rats. The in utero treated rats also responded with a lower DA release to amphetamine (1 mg/kg ip) administration. On the other hand, no difference in NAC DA level was found amongst treatment groups in response to different concentrations of the D(2)-D(3) dopaminergic agonist quinpirole, locally administered by means of inverse dialysis. These data indicate a preferential impairment of NAC DA synthesis and/or release in rats exposed to Pb acetate during their intrauterine life.


Asunto(s)
Dopamina/metabolismo , Plomo/toxicidad , Núcleo Accumbens/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dopamina/fisiología , Femenino , Lactancia/metabolismo , Plomo/sangre , Plomo/farmacocinética , Masculino , Microdiálisis , Núcleo Accumbens/metabolismo , Embarazo , Ratas , Ratas Long-Evans
17.
Behav Brain Res ; 118(1): 61-5, 2001 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-11163634

RESUMEN

The rewarding effects of morphine, cocaine, amphetamine and nicotine were evaluated in CB1 receptor knockout mice by means of an intravenous self-administration model. Experiments were carried out on drug-naive animals using a nose-poking response (NPR)-like as operandum. The results of the present study indicate that morphine did not induce intravenous self-administration in mutant CB1 receptor knockout mice, whereas it was significantly self-administered by the corresponding wild type mice. On the contrary, cocaine, amphetamine and nicotine were self-administered to the same extent by both wild type and CB1 receptor knockout mice. These data clearly indicate that the CB1 cannabinoid receptor is essential not only for the expression of cannabinoid reinforcing effects but also for the modulation of morphine rewarding effects. The specificity of such interaction is supported by the finding that contrary to morphine, cocaine, d-amphetamine and nicotine were self-administered by mice at the same extent either in presence or in absence of the CB1 receptor.


Asunto(s)
Conducta Adictiva/metabolismo , Cannabinoides/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Narcóticos/farmacología , Agonistas Nicotínicos/farmacología , Receptores de Droga/efectos de los fármacos , Anfetamina/administración & dosificación , Anfetamina/farmacología , Animales , Cocaína/administración & dosificación , Cocaína/farmacología , Masculino , Ratones , Ratones Noqueados , Morfina/administración & dosificación , Morfina/farmacología , Narcóticos/administración & dosificación , Nicotina/administración & dosificación , Nicotina/farmacología , Receptores de Cannabinoides , Receptores de Droga/metabolismo , Recompensa
18.
Eur J Pharmacol ; 406(1): 75-7, 2000 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-11011036

RESUMEN

Two cyclo-oxygenase inhibitors, indomethacin and nimesulide, have been shown to potentiate morphine-induced stimulation of meso-accumbens dopamine neurons. In this study, an unbiased conditioned place preference procedure was used to evaluate whether nimesulide produces motivational effect after systemic administration in rats. These results show that nimesulide, at doses 0.1, 0.5 and 1 mg/kg, even lower than those usually applied for inflammatory conditions, induces conditioned place preference in rats, suggesting that it might possess rewarding properties in humans.


Asunto(s)
Condicionamiento Psicológico/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Sulfonamidas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Morfina/farmacología , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
19.
Alcohol ; 20(3): 247-56, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10869866

RESUMEN

Gamma-hydroxybutyric acid, an endogenous compound present in mammalian brain and supposed to be a neurotransmitter or neuromodulator, has been shown to affect several aspects of dependence from some drugs of abuse. It has been successfully used in clinical practice to alleviate both alcohol and opiate withdrawal symptoms. The aim of this study was to investigate whether gamma-hydroxybutyric acid possesses rewarding properties by means of conditioned place preference and intravenous self-administration paradigms. In the present study, gamma-hydroxybutyric acid induced conditioned place preference in rats, was intravenously self-administered by drug-naive mice, and altered cocaine intravenous self-administration in rats. Although to date the physiological role of this compound still remains unclear, there is no doubt that gamma-hydroxybutyric acid, in addition to its proved effect on alcohol and opiate dependence, possesses reinforcing properties of its own and may interfere with the neurochemical events in the rewarding effects produced by psychostimulant drugs. Our investigation points out the abuse liability of this drug, suggesting the use of particular precaution in handling gamma-hydroxybutyric acid as a clinically useful drug.


Asunto(s)
Hidroxibutiratos , Recompensa , Animales , Cocaína/administración & dosificación , Condicionamiento Psicológico , Hidroxibutiratos/administración & dosificación , Hidroxibutiratos/uso terapéutico , Inyecciones Intravenosas , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración , Trastornos Relacionados con Sustancias
20.
Eur J Pharmacol ; 383(3): R1-2, 1999 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-10594337

RESUMEN

Morphine (10 and 20 mg/kg, s.c.) does not modify dopamine release in the nucleus accumbens of cannabinoid CB(1) knock-out mice under conditions where it dose-dependently stimulates the release of dopamine in the corresponding wild-type mice. These results demonstrate that cannabinoid CB(1) receptors, regulate mesolimbic dopaminergic transmission in brain areas known to be involved in the reinforcing effects of morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Dopamina/metabolismo , Morfina/farmacología , Núcleo Accumbens/efectos de los fármacos , Receptores de Droga/efectos de los fármacos , Animales , Ratones , Ratones Noqueados , Núcleo Accumbens/metabolismo , Receptores de Cannabinoides , Receptores de Droga/genética , Receptores de Droga/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA