Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Neurobiol ; 23(3): 430-5, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23642859

RESUMO

Optogenetic tools have revolutionized the field of neuroscience, and brought the study of neural circuits to a higher level. Optogenetics has significantly improved our understanding not only of the neuronal connections and function of the healthy brain, but also of the neuronal changes that lead to psychiatric disorders. In this review, we summarize recent optogenetic studies that explored different brain circuits involved in natural behaviors, such as sleep and arousal, reward, fear, and social and aggressive behavior. In addition, we describe how alterations in these circuits may lead to psychiatric disorders such as addiction, anxiety, depression, or schizophrenia.


Assuntos
Encéfalo/fisiopatologia , Transtornos Mentais , Optogenética/métodos , Animais , Humanos , Transtornos Mentais/diagnóstico , Transtornos Mentais/fisiopatologia , Transtornos Mentais/terapia , Optogenética/tendências
2.
Eur J Pharmacol ; 711(1-3): 63-72, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23632394

RESUMO

While opioids are potent analgesics widely used in the management of pain, a number of well-known adverse effects limit their use. The sigma-1 receptor is a ligand-regulated molecular chaperone involved in pain processing, including modulation of opioid antinociception. However, data supporting the potential use of sigma-1 receptor ligands as suitable opioid adjuvants are based on studies that use non selective ligands. Also, safety issues derived from combination therapy are poorly addressed. In this study we used the new selective sigma-1 receptor antagonist S1RA (E-52862) to characterize the effect of selective sigma-1 receptor blockade on opioid-induced efficacy- and safety-related outcomes in mice. S1RA (40 mg/kg) had no effect in the tail-flick test but did enhance the antinociceptive potency of several opioids by a factor between 2 and 3.3. The potentiating effect of S1RA on morphine antinociception did not occur in sigma-1 receptor knockout mice, which supports the selective involvement of the sigma-1 receptor. Interestingly, S1RA co-administration restored morphine antinociception in tolerant mice and reverted the reward effects of morphine in the conditioned place preference paradigm. In addition, enhancement of antinociception was not accompanied by potentiation of other opioid-induced effects, such as the development of morphine analgesic tolerance, physical dependence, inhibition of gastrointestinal transit, or mydriasis. The use of sigma-1 receptor antagonists as opioid adjuvants could represent a promising pharmacological strategy to enhance opioid potency and, most importantly, to increase the safety margin of opioids. S1RA is currently in phase II clinical trials for the treatment of several pain conditions.


Assuntos
Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Receptores sigma/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Quimioterapia Adjuvante , Condicionamento Psicológico/efeitos dos fármacos , Sinergismo Farmacológico , Tolerância a Medicamentos , Trânsito Gastrointestinal/efeitos dos fármacos , Técnicas de Inativação de Genes , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Masculino , Camundongos , Morfina/efeitos adversos , Morfina/farmacologia , Midríase/induzido quimicamente , Naloxona/farmacologia , Receptores sigma/deficiência , Receptores sigma/genética , Recompensa , Comportamento Espacial/efeitos dos fármacos , Receptor Sigma-1
3.
J Neurosci ; 33(18): 7618-26, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23637156

RESUMO

The lateral hypothalamus (LH) sends a dense glutamatergic and peptidergic projection to dopamine neurons in the ventral tegmental area (VTA), a cell group known to promote reinforcement and aspects of reward. The role of the LH to VTA projection in reward-seeking behavior can be informed by using optogenetic techniques to dissociate the actions of LH neurons from those of other descending forebrain inputs to the VTA. In the present study, we identify the effect of neurotensin (NT), one of the most abundant peptides in the LH to VTA projection, on excitatory synaptic transmission in the VTA and reward-seeking behavior. Mice displayed robust intracranial self-stimulation of LH to VTA fibers, an operant behavior mediated by NT 1 receptors (Nts1) and NMDA receptors. Whole-cell patch-clamp recordings of VTA dopamine neurons demonstrated that NT (10 nm) potentiated NMDA-mediated EPSCs via Nts1. Results suggest that NT release from the LH into the VTA activates Nts1, thereby potentiating NMDA-mediated EPSCs and promoting reward. The striking behavioral and electrophysiological effects of NT and glutamate highlight the LH to VTA pathway as an important component of reward.


Assuntos
Condicionamento Operante/fisiologia , Ácido Glutâmico/metabolismo , Hipotálamo/fisiologia , Neurotensina/metabolismo , Recompensa , Área Tegmentar Ventral/fisiologia , Animais , Proteínas de Bactérias/genética , Channelrhodopsins , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Hipotálamo/efeitos dos fármacos , Técnicas In Vitro , Luz , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Vias Neurais/fisiologia , Neurotensina/farmacologia , Pirazóis/farmacologia , Quinolinas/farmacologia , Quinoxalinas/farmacologia , Receptores de Neurotensina/antagonistas & inibidores , Receptores de Neurotensina/deficiência , Autoestimulação , Transdução de Sinais/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo , Valina/análogos & derivados , Valina/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos
4.
Br J Pharmacol ; 167(4): 892-904, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22612385

RESUMO

BACKGROUND AND PURPOSE: The modulatory activity of the orphan receptor GPR3 in the brain has been related to the control of emotional behaviours. Limbic structures that express GPR3 have been associated with the effects of drug abuse. EXPERIMENTAL APPROACH: The role of GPR3 in different cocaine-elicited behaviours including locomotor activity, behavioural sensitization, conditioned place preference (CPP) and intravenous self-administration was evaluated in Gpr3-/- mice and their Gpr3+/+ littermates. Cocaine-induced dopamine release in the nucleus accumbens was also evaluated to elucidate the effect of Gpr3 deletion on extracellular levels of dopamine. KEY RESULTS: Gpr3-/- mice exhibited higher rewarding responses in the CPP paradigm. Gpr3-/- mice self-administered more cocaine, especially during the first days of training. No differences were found between genotypes regarding behavioural sensitization and the maximal effort required to obtain a cocaine infusion. Non-contingent priming injections of cocaine before operant training eliminated enhanced cocaine self-administration in Gpr3-/- mice. Extracellular levels of dopamine in the nucleus accumbens induced by cocaine did not differ between genotypes. CONCLUSIONS AND IMPLICATIONS: The increased responsiveness of Gpr3-/- mice to the acute locomotor effects of cocaine and the inconsistency to further increase this effect reflected an 'already maximally sensitized' basal state. Enhanced responsiveness of Gpr3-/- mice to cocaine reward and to early phases of reinforcement suggests that an initial alteration increased vulnerability to this type of drug abuse. Overall, altered signalling pathways of GPR3 could contribute to the neurobiological substrate involved in developing addiction to cocaine.


Assuntos
Cocaína/administração & dosagem , Receptores Acoplados a Proteínas G/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante , Dopamina/metabolismo , Alimentos , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Reforço Psicológico , Autoadministração
5.
J Neurosci ; 31(30): 10829-35, 2011 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-21795535

RESUMO

Phasic activation of dopaminergic neurons is associated with reward-predicting cues and supports learning during behavioral adaptation. While noncontingent activation of dopaminergic neurons in the ventral tegmental are (VTA) is sufficient for passive behavioral conditioning, it remains unknown whether the phasic dopaminergic signal is truly reinforcing. In this study, we first targeted the expression of channelrhodopsin-2 to dopaminergic neurons of the VTA and optimized optogenetically evoked dopamine transients. Second, we showed that phasic activation of dopaminergic neurons in freely moving mice causally enhances positive reinforcing actions in a food-seeking operant task. Interestingly, such effect was not found in the absence of food reward. We further found that phasic activation of dopaminergic neurons is sufficient to reactivate previously extinguished food-seeking behavior in the absence of external cues. This was also confirmed using a single-session reversal paradigm. Collectively, these data suggest that activation of dopaminergic neurons facilitates the development of positive reinforcement during reward-seeking and behavioral flexibility.


Assuntos
Condicionamento Operante/fisiologia , Dopamina/metabolismo , Estimulação Luminosa , Recompensa , Área Tegmentar Ventral/fisiologia , Análise de Variância , Animais , Proteínas de Bactérias/genética , Channelrhodopsins , Discriminação Psicológica , Estimulação Elétrica/métodos , Técnicas Eletroquímicas , Regulação da Expressão Gênica/fisiologia , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/fisiologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/genética , Área Tegmentar Ventral/citologia
6.
PLoS One ; 5(2): e9143, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-20174577

RESUMO

The majority of MDMA (ecstasy) recreational users also consume cannabis. Despite the rewarding effects that both drugs have, they induce several opposite pharmacological responses. MDMA causes hyperthermia, oxidative stress and neuronal damage, especially at warm ambient temperature. However, THC, the main psychoactive compound of cannabis, produces hypothermic, anti-inflammatory and antioxidant effects. Therefore, THC may have a neuroprotective effect against MDMA-induced neurotoxicity. Mice receiving a neurotoxic regimen of MDMA (20 mg/kg x 4) were pretreated with THC (3 mg/kg x 4) at room (21 degrees C) and at warm (26 degrees C) temperature, and body temperature, striatal glial activation and DA terminal loss were assessed. To find out the mechanisms by which THC may prevent MDMA hyperthermia and neurotoxicity, the same procedure was carried out in animals pretreated with the CB(1) receptor antagonist AM251 and the CB(2) receptor antagonist AM630, as well as in CB(1), CB(2) and CB(1)/CB(2) deficient mice. THC prevented MDMA-induced-hyperthermia and glial activation in animals housed at both room and warm temperature. Surprisingly, MDMA-induced DA terminal loss was only observed in animals housed at warm but not at room temperature, and this neurotoxic effect was reversed by THC administration. However, THC did not prevent MDMA-induced hyperthermia, glial activation, and DA terminal loss in animals treated with the CB(1) receptor antagonist AM251, neither in CB(1) and CB(1)/CB(2) knockout mice. On the other hand, THC prevented MDMA-induced hyperthermia and DA terminal loss, but only partially suppressed glial activation in animals treated with the CB(2) cannabinoid antagonist and in CB(2) knockout animals. Our results indicate that THC protects against MDMA neurotoxicity, and suggest that these neuroprotective actions are primarily mediated by the reduction of hyperthermia through the activation of CB(1) receptor, although CB(2) receptors may also contribute to attenuate neuroinflammation in this process.


Assuntos
Dronabinol/farmacologia , Febre/prevenção & controle , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Síndromes Neurotóxicas/prevenção & controle , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Temperatura Corporal/efeitos dos fármacos , Febre/induzido quimicamente , Alucinógenos/toxicidade , Indóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Síndromes Neurotóxicas/etiologia , Piperidinas/farmacologia , Psicotrópicos/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/metabolismo , Temperatura
7.
Biol Psychiatry ; 63(11): 1030-8, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17950256

RESUMO

BACKGROUND: 3,4-Methylenedioxymethamphetamine (MDMA) is a popular recreational drug widely abused by young people. The endocannabinoid system is involved in the addictive processes induced by different drugs of abuse. However, the role of this system in the pharmacological effects of MDMA has not yet been clarified. METHODS: Locomotion, body temperature, and anxiogenic-like responses were evaluated after acute MDMA administration in CB(1) cannabinoid receptor 1 knockout mice. Additionally, MDMA rewarding properties were investigated in the place conditioning and the intravenous self-administration paradigms. Extracellular levels of dopamine (DA) in the nucleus accumbens were also analyzed after a single administration of MDMA by in vivo microdialysis. RESULTS: Acute MDMA administration increased locomotor activity, body temperature, and anxiogenic-like responses in wild-type mice, but these responses were lower or abolished in knockout animals. 3,4-Methylenedioxymethamphetamine produced similar conditioned place preference and increased dopamine extracellular levels in the nucleus accumbens in both genotypes. Nevertheless, CB(1) knockout mice failed to self-administer MDMA at any of the doses used. CONCLUSIONS: These results indicate that CB(1) cannabinoid receptors play an important role in the acute prototypical effects of MDMA and are essential in the acquisition of an operant behavior to self-administer this drug.


Assuntos
3,4-Metilenodioxianfetamina/farmacologia , Condicionamento Operante/efeitos dos fármacos , Alucinógenos/farmacologia , Receptor CB1 de Canabinoide/fisiologia , Reforço Psicológico , Análise de Variância , Animais , Ansiedade/induzido quimicamente , Comportamento Animal/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Masculino , Camundongos , Camundongos Knockout , Microdiálise/métodos , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptor CB1 de Canabinoide/deficiência , Autoadministração
8.
Psychopharmacology (Berl) ; 193(1): 75-84, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17387458

RESUMO

INTRODUCTION: 3, 4-Methylenedioxymethamphetamine (MDMA) and cannabis are widely abused illicit drugs that are frequently consumed in combination. Interactions between these two drugs have been reported in several pharmacological responses observed in animals, such as body temperature, anxiety, cognition, and reward. However, the interaction between MDMA and cannabis in addictive processes such as physical dependence has not been elucidated yet. DISCUSSION: In this study, the effects of acute and chronic MDMA were evaluated on the behavioral manifestations of Delta(9)-tetrahydrocannabinol (THC) abstinence in mice. THC withdrawal syndrome was precipitated by injecting the cannabinoid antagonist rimonabant (10 mg/kg, i.p.) in mice chronically treated with THC and receiving MDMA (2.5, 5 and 10 mg/kg i.p.) or saline just before the withdrawal induction or chronically after the THC administration. RESULTS: Both chronic and acute MDMA decreased in a dose-dependent manner the severity of THC withdrawal. In vivo microdialysis experiments showed that acute MDMA (5 mg/kg, i.p.) administration increased extracellular serotonin levels in the prefrontal cortex, but not dopamine levels in the nucleus accumbens. Our results also indicate that the attenuation of THC abstinence symptoms was not due to a direct interaction between rimonabant and MDMA nor to the result of the locomotor stimulating effects of MDMA. CONCLUSION: The modulation of the cannabinoid withdrawal syndrome by acute or chronic MDMA suggests a possible mechanism to explain the associated consumption of these two drugs in humans.


Assuntos
Dronabinol/efeitos adversos , N-Metil-3,4-Metilenodioxianfetamina/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Antagonistas de Receptores de Canabinoides , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos , Atividade Motora/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Piperidinas/farmacologia , Pirazóis/farmacologia , Rimonabanto , Serotonina/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia
9.
Neuropsychopharmacology ; 30(9): 1670-80, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15742004

RESUMO

Acute rewarding properties are essential for the establishment of cocaine addiction, and multiple neurochemical processes participate in this complex behavior. In the present study, we used the self-administration paradigm to evaluate the role of CB1 cannabinoid receptors in several aspects of cocaine reward, including acquisition, maintenance, and motivation to seek the drug. For this purpose, both CB1 receptor knockout mice and wild-type littermates were trained to intravenously self-administer cocaine under different schedules. Several cocaine training doses (0.32, 1, and 3.2 mg/kg/infusion) were used in the acquisition studies. Only 25% of CB1 knockout mice vs 75% of their wild-type littermates acquired a reliable operant responding to self-administer the most effective dose of cocaine (1 mg/kg/infusion), and the number of sessions required to attain this behavior was increased in knockout mice. Animals reaching the acquisition criteria were evaluated for the motivational strength of cocaine as a reinforcer under a progressive ratio schedule. The maximal effort to obtain a cocaine infusion was significantly reduced after the genetic ablation of CB1 receptors. A similar result was obtained after the pharmacological blockade of CB1 receptors with SR141716A in wild-type mice. Moreover, the cocaine dose-response curve was flattened in the knockout group, suggesting that the differences observed between genotypes were related to changes in the reinforcing efficacy of the training dose of cocaine. Self-administration for water and food was not altered in CB1 knockout mice in any of the reinforcement schedules used, which emphasizes the selective impairment of drug reinforcement in these knockout mice. Finally, cocaine effects on mesolimbic dopaminergic transmission were evaluated by in vivo microdialysis in these mice. Acute cocaine administration induced a similar enhancement in the extracellular levels of dopamine in the nucleus accumbens of both CB1 knockout and wild-type mice. This work clearly demonstrates that CB1 receptors play an important role in the consolidation of cocaine reinforcement, although are not required for its acute effects on mesolimbic dopaminergic transmission.


Assuntos
Anestésicos Locais/administração & dosagem , Cocaína/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Receptor CB1 de Canabinoide/deficiência , Autoadministração , Análise de Variância , Animais , Comportamento Animal/fisiologia , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Comportamento Alimentar/fisiologia , Privação de Alimentos/fisiologia , Camundongos , Camundongos Knockout , Microdiálise/métodos , Piperidinas/administração & dosagem , Pirazóis/administração & dosagem , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto , Fatores de Tempo , Privação de Água/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...