Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.177
Filtrar
1.
Pharmacol Biochem Behav ; 213: 173333, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35065952

RESUMO

Dissociative-based new psychoactive substances (NPSs) are increasingly available through the Internet, and public health problems related to the recreational use of these substances have been increasing globally. Two such NPSs are deschloroketamine and diphenidine, which are primarily used recreationally as ketamine substitutes. However, there is little scientific evidence to describe the dependence liability of NPSs. This study aimed to evaluate the dependence liability of deschloroketamine and diphenidine via animal behavioral experiments. We evaluated the rewarding and reinforcing effects of these NPSs using the conditioned place preference (CPP) and the self-administration (SA) paradigms in mice. Psychomotor effects and behavioral features of these compounds were assessed by quantifying locomotor activity, stereotypic movements, and dopaminergic neurotransmission. Both deschloroketamine (10 mg/kg) and diphenidine (10-60 mg/kg) produced increased locomotor activation and stereotypy that were similar to the effects of ketamine (10 mg/kg). Both deschloroketamine (10 mg/kg) and diphenidine (10, 20 mg/kg) increased the animals' preference for the drug-paired compartment in the CPP testing. In the SA testing, deschloroketamine (1 mg/kg/infusion) increased the number of active lever presses and the number of infusions received, whereas diphenidine administration (1, 2 mg/kg/infusion) did not alter either of these. Furthermore, both deschloroketamine and diphenidine increased dopamine levels in PC-12 cells. Collectively, the data suggest that deschloroketamine may have both rewarding and reinforcing effects, whereas diphenidine only induced rewarding effect.


Assuntos
Ketamina/farmacologia , Piperidinas/farmacologia , Reforço Psicológico , Recompensa , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Dopamina/metabolismo , Ketamina/administração & dosagem , Ketamina/análogos & derivados , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Piperidinas/administração & dosagem , Psicotrópicos/farmacologia , Ratos , Autoadministração , Comportamento Estereotipado/efeitos dos fármacos
2.
Behav Brain Res ; 422: 113759, 2022 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-35051488

RESUMO

Conditioned avoidance responses (CAR) behavior is a classical instrumental response paradigm, which is widely used to study aversive conditioning and defensive motivation behavior. Previous studies have shown that dopamine D1 and D2 receptors are involved in CAR behavior; however, it is unclear in which brain regions that dopamine evokes CAR behavior. The aim of the study is to investigate whether dopamine triggers CAR behavior via activating dopamine D1 or D2 receptors in the shell of nucleus accumbens or dorsolateral striatum. The present study found that infusion of the dopamine D2 receptor agonist quinpirole, but not D1 receptor agonist SKF38393, into the shell of nucleus accumbens evoked CAR behavior in reserpine-treated rats. Whereas, infusion of neither SKF38393 nor quinpirole into the dorsolateral striatum evoked CAR behavior. In addition, infusion of quinpirole into the shell of nucleus accumbens enhanced CAR behavior in the unsuccessful trained rats without affecting the motor function in the balance beam and locomotor tests. In conclusion, activation of dopamine D2, but not D1 receptors in the shell of nucleus accumbens evokes CAR behavior. However, activation of dopamine D1 and D2 receptors in the dorsolateral striatum does not evoke CAR behavior. It is suggested that the shell of nucleus accumbens is the critical brain region for dopamine to invoke CAR behavior, and activation of dopamine D2 receptors in the shell of nucleus accumbens is sufficient and necessary to evoke CAR behavior.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Animais , Masculino , Neostriado/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/efeitos dos fármacos
3.
Behav Brain Res ; 421: 113713, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-34906607

RESUMO

Naphyrone, also known as NRG-1, is a novel psychoactive substance (NPS), a cathinone with stimulatory properties available on the grey/illicit drug market for almost a decade. It is structurally related to infamously known powerful stimulants with the pyrovalerone structure, such as alpha-pyrrolidinovalerophenone (α-PVP) or methylenedioxypyrovalerone (MDPV) that are labeled as a cheap replacement for cocaine and other stimulants. Despite the known addictive potential of α-PVP and MDPV, there are no studies directly evaluating naphyrone's addictive potential e.g., in conditioned place preference (CPP) test or using self-administration. Therefore, our study was designed to evaluate the addictive potential in a CPP test in male Wistar rats and compare its effect to another powerful stimulant with a high addictive potential - methamphetamine. Naphyrone increased time spent in the drug-paired compartment with 5 and 20 mg/kg s.c. being significant and 10 mg/kg s.c. reaching the threshold (p = 0.07); the effect was comparable to that of methamphetamine 1.5 mg/kg s.c. The lowest dose, naphyrone 1 mg/kg s.c., had no effect on CPP. Interestingly, no dose response effect was detected. Based on these data, we are able to conclude that naphyrone has an addictive potential and may possess a significant risk to users.


Assuntos
Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Metanfetamina/farmacologia , Pentanonas/farmacologia , Pirrolidinas/farmacologia , Transtornos Relacionados ao Uso de Substâncias , Alcaloides/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Metanfetamina/administração & dosagem , Pentanonas/administração & dosagem , Pirrolidinas/administração & dosagem , Ratos , Ratos Wistar
4.
Artigo em Inglês | MEDLINE | ID: mdl-34929324

RESUMO

Drug-related memory can be transiently destabilized by memory retrieval, after which memories are reconsolidated. Neurons in the basolateral amygdala (BLA) that are activated by emotional information may be one of the key mechanisms underlying this destabilization. However, the specific neural circuits underlying this destabilization process remain unknown. Because BLA receives noradrenergic inputs from the nucleus tractus solitarius (NTS) and locus coeruleus (LC), we studied the role of afferent projections into the BLA in the destabilization of morphine self-administration memory in rats. We first showed that morphine (unconditioned stimulus, US) + morphine-associated conditioned stimuli (CS) exposure, rather than CS exposure alone, destabilized morphine self-administration memory. Then, we measured projection-specific activation after the US + CS or CS retrieval test using c-fos (activity marker)-labeling in projection areas. Compared with CS exposure, we found that US + CS exposure induced more neuronal activation in the BLA and NTS but not in the LC. Next, we determined the effects of chemogenetic inactivation or activation of NTS or LC projections to BLA (NTS â†’ BLA or LC â†’ BLA) on this destabilization. We found that NTS â†’ BLA, but not LC â†’ BLA inactivation during memory retrieval, prevented memory destabilization induced by US + CS exposure. Furthermore, NTS â†’ BLA, but not LC â†’ BLA activation during CS retrieval induced destabilization. Thus, our results identify a specific neural circuit underlying the transformation of a stable opiate-associated memory into an unstable memory and subsequently guide reconsolidation.


Assuntos
Analgésicos Opioides/farmacologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Locus Cerúleo/fisiologia , Memória/efeitos dos fármacos , Morfina/farmacologia , Núcleo Solitário/fisiologia , Animais , Condicionamento Clássico/efeitos dos fármacos , Masculino , Norepinefrina , Ratos , Autoadministração
5.
Brain Res Bull ; 179: 36-48, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34871711

RESUMO

Methamphetamine (METH), a synthetically produced central nervous system stimulant, is one of the most illicit and addictive drugs worldwide. Protein phosphatase Mg2 + /Mn2 + -dependent 1F F (PPM1F) has been reported to exert multiple biological and cellular functions. Nevertheless, the effects of PPM1F and its neuronal substrates on METH addiction remain unclear. Herein, we first established a METH-induced conditioned place preference (CPP) mouse model. We showed that PPM1F is widely distributed in 5-HT neurons of the dorsal raphe nucleus (DRN), and METH treatment decreased the expression of PPM1F in DRN, which was negatively correlated with METH-induced CPP behaviors. Knockout of PPM1F mediated by adeno-associated virus (AAV) in DRN produced enhanced susceptibility to METH-induced CPP, whereas the overexpression of PPM1F in DRN attenuated METH-induced CPP phenotypes. The expression levels of Tryptophan hydroxylase2 (TPH2) and serotonin transporter (SERT) were down-regulated with a concurrent reduction in 5-hydroxytryptamine (5-HT), tryptophan hydroxylase2 (TPH2)-immunoreactivity neurons and 5-HT levels in DRN of PPM1F knockout mice. In the end, decreased expression levels of PPM1F were found in the blood of METH abusers and METH-taking mice. These results suggest that PPM1F in DRN 5-HT neurons regulates METH-induced CPP behaviors by modulating the key components of the 5-HT neurotransmitter system, which might be an important pathological gene and diagnostic marker for METH-induced addiction.


Assuntos
Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Metanfetamina/farmacologia , Fosfoproteínas Fosfatases/efeitos dos fármacos , Neurônios Serotoninérgicos/efeitos dos fármacos , Animais , Condicionamento Clássico/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/metabolismo
6.
Neurosci Lett ; 768: 136383, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34864087

RESUMO

BACKGROUND: The flavonoid quercetin has several pharmacological effects on the nervous system. Previous research showed that quercetin has useful influences on some mechanisms that are relevant in drug and substance addiction. Alcohol addiction, also known as alcoholism, is a disorder that influences the population in all walks of life. The purpose of the current study was to investigate whether quercetin affects the acquisition, extinction, and reinstatement of ethanol-induced conditioned place preference (ethanol-CPP) in adolescent mice. METHODS: CPP was established by administration of intraperitoneal (i.p.) ethanol (2.0 g/kg) in a conditioning trial. The mice were pretreated with quercetin (at doses of 10, 30, and 100 mg/kg, i.p.) 30 minutes before each ethanol injection to test the effects of quercetin on the reward properties of ethanol. Ethanol-CPP was extinguished (13-days) by repeated testing, during which conditioned mice were given different doses of quercetin every day. Lastly, efficacy of quercetin in preventing reinstatement of ethanol-CPP triggers was also assessed by the administration of single dose ethanol (0.4 g/kg, i.p.). RESULTS: Quercetin pretreatment attenuated the acquisition and reinstatement. In addition, quercetin administration accelerated the extinction of ethanol-CPP. CONCLUSIONS: In conclusion, these results may cast a novel light on quercetin as an agent that could be potentially useful to attenuate different effects of ethanol and as adjuvant pharmacotherapy for ethanol addiction. However, future studies are needed to demonstrate the detailed underlying mechanisms of quercetin on ethanol addiction.


Assuntos
Alcoolismo , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Quercetina/farmacologia , Recompensa , Animais , Condicionamento Clássico/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Masculino , Camundongos
7.
Behav Brain Res ; 416: 113537, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34416299

RESUMO

Cocaine use disorder is a serious, chronic and relapsing disease of the nervous system, for which effective treatments do not yet exist. Recently, the role of the N-methyl-d-aspartate (NMDA) receptor subunit GluN2B has been highlighted in cocaine abstinence followed by extinction training. Since the GluN2B subunit is stabilized at synaptic level by the interaction with its scaffolding protein PSD95, in this study we aimed at investigating efficacy of Tat-NR2B9c peptide, a PSD95 inhibitor, which disrupts the interaction of PSD95 with GluN2B, in the attenuation of cocaine seeking-behavior or cue-induced reinstatement. We found that Tat-NR2B9c, administered intravenously, attenuated the reinstatement of active lever presses induced by a priming dose of cocaine or by drug-associated conditioned stimuli. At the same time, the GluN2B/PSD95 complex levels were decreased in the ventral hippocampus of rats that previously self-administered cocaine injected with Tat-NR2B9c during cocaine- or cue-induced reinstatement. In conclusion, we here provide the first evidence showing that the disruption of the GluN2B/PSD95 complexes during cocaine abstinence followed by extinction training may represent a useful strategy to reduce reinstatement of cocaine-seeking behavior.


Assuntos
Cocaína/farmacologia , Comportamento de Procura de Droga , Extinção Psicológica/fisiologia , Peptídeos/antagonistas & inibidores , Autoadministração , Administração Intravenosa , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Sinais (Psicologia) , Proteína 4 Homóloga a Disks-Large/metabolismo , Masculino , Peptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley
8.
Behav Brain Res ; 416: 113579, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34508768

RESUMO

Natural rewards and abused drugs affect the function of the common brain's reward system. Interaction between social and drug rewards can change the vulnerability to development of drug addiction. Here, we investigate the effects of sexual experience and sex deprivation on the acquisition, maintenance, and drug prime-induced reinstatement of morphine-seeking behavior in male mice using conditioned place preference (CPP). CPP induced with morphine (3, 5, 7 mg/kg, s.c. for 3 days) lasted for 10 days after cessation of morphine treatment and priming dose of morphine (2 mg/kg, s.c.) reinstated the extinguished CPP. In the post-test phase, sexually experienced animals showed a lower preference for morphine compared to sex-deprived males. In the extinction phase, sex deprivation shortened maintenance time compared to control animals. The preference for morphine in sexually experienced animals did not diminish by the seventeenth extinction day. In both groups, the priming injection of morphine after the extinction period could reinstate the extinguished morphine-induced CPP. Together, these data showed the interaction between sex and drug reward and that sexual behavior -a natural rewarding stimulus- can prolong, whereas sex deprivation can block the maintenance of morphine-seeking behaviors. Sexual experience may induce functional and morphological alterations in brain reward areas particularly the mesolimbic system similar to repeated exposure to abused drugs which can affect morphine-seeking behaviors.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Morfina/farmacologia , Recompensa , Comportamento Sexual/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Camundongos , Dependência de Morfina , Transtornos Relacionados ao Uso de Substâncias
9.
Artigo em Inglês | MEDLINE | ID: mdl-34509531

RESUMO

Opioid withdrawal can be associated to environmental cues through classical conditioning. Exposure to these cues can precipitate a state of conditioned withdrawal in abstinent subjects, and there are suggestions that conditioned withdrawal can perpetuate the addiction cycle in part by promoting the storage of memories. This review discusses evidence supporting the hypothesis that conditioned withdrawal facilitates memory consolidation by activating a neurocircuitry that involves the extended amygdala. Specifically, the central amygdala, the bed nucleus of the stria terminalis, and the nucleus accumbens shell interact functionally during withdrawal, mediate expression of conditioned responses, and are implicated in memory consolidation. From this perspective, the extended amygdala could be a neural pathway by which drug-seeking behaviour performed during a state of conditioned withdrawal is more likely to become habitual and persistent.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Condicionamento Clássico/efeitos dos fármacos , Sinais (Psicologia) , Consolidação da Memória/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Comportamento Aditivo/fisiopatologia , Comportamento de Procura de Droga , Humanos , Vias Neurais , Núcleo Accumbens/fisiopatologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ratos
10.
Pharmacol Biochem Behav ; 211: 173286, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34634300

RESUMO

RATIONALE: Exposure to a drug can subsequently impact its own reactivity as well as that of other drugs. Given that users of synthetic cathinones, i.e., "bath salts", typically have extensive and varied drug histories, an understanding of the effects of drug history on the behavioral and physiological consequences of synthetic cathiones may be important to their abuse liability. OBJECTIVES: The goal of the current work was to assess the effects of an ethanol pre-exposure on the rewarding and aversive effects of α-PVP. METHODS: Adult male Sprague Dawley rats were exposed to ethanol prior to combined conditioned taste avoidance/conditioned place preference training in which rats were injected with 1.5, 3 or 5 mg/kg of racemic α-PVP or vehicle. Following a 7-day washout period, rats were then tested for thermoregulatory effects of α-PVP using subcutaneous probes to measure body temperature changes over the course of 8 h. This was followed 10 days later by assessments for α-PVP-induced locomotor activity and stereotypies over a 1-h session. RESULTS: α-PVP induced significant dose- and trial-dependent taste avoidance that was significantly attenuated by ethanol history and dose- and time-dependent increases in locomotor activity that were significantly increased by ethanol. α-PVP also induced place preferences and dose- and time-dependent increases in body temperature, but these measures were unaffected by ethanol history. CONCLUSIONS: α-PVP's aversive effects (as measured by taste avoidance) were attenuated, while its rewarding effects (as indexed by place preference conditioning) were unaffected, by ethanol pre-exposure. Such a pattern may indicate increased α-PVP abuse liability, as changes in the balance of aversion and reward may impact overall drug effects and likelihood of drug intake. Future self-administration studies will be necessary to explore this possibility.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Etanol/farmacologia , Pentanonas/farmacologia , Pirrolidinas/farmacologia , Recompensa , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Alcaloides/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Locomoção/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Autoadministração , Paladar/efeitos dos fármacos
11.
Neurobiol Learn Mem ; 185: 107528, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34607024

RESUMO

Contextual fear conditioning, where the prevailing situational cues become associated with an aversive unconditional stimulus such as electric shock, is sexually dimorphic. Males typically show higher levels of fear than females. There are two components to contextual fear conditioning. First the multiple cues that encompass the context must be integrated into a coherent representation, a process that requires the hippocampus. The second is that representation must be communicated to the basolateral amygdala where it can be associated with shock. If there is inadequate time for forming the representation prior to shock poor conditioning results and this is called the immediate shock deficit. One can isolate the contextual processing component, as well as alleviate the deficit, by providing an opportunity to explore the context without shock prior to the conditioning session. The purpose of the present study was to determine the extent to which cholinergic processes within the dentate gyrus of the hippocampus during contextual processing contribute to the sexual dimorphism. Clozapine-n-oxide (CNO) is a putatively inactive compound that acts only upon synthetic genetically engineered receptors. However, we found that CNO infused into the dentate gyrus prior to exploration eliminated the sexual dimorphism by selectively decreasing freezing in males to the level of females. Biological activity of CNO is usually attributed to metabolism of CNO to clozapine and we found that clozapine, and the muscarinic cholinergic antagonist, scopolamine, produced results similar to CNO, preferentially affecting males. On the other hand, the muscarinic agonist oxotremorine selectively impaired conditioning in females. Overall, the current experiments reveal significant off-target effects of CNO and implicate muscarinic cholinergic receptors in the dentate gyrus as a significant mediator of the sexual dimorphism in contextual fear conditioning.


Assuntos
Condicionamento Clássico/fisiologia , Giro Denteado/fisiologia , Medo/fisiologia , Animais , Clozapina/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Giro Denteado/anatomia & histologia , Giro Denteado/efeitos dos fármacos , Feminino , Masculino , Ratos , Ratos Long-Evans , Escopolamina/farmacologia , Caracteres Sexuais
12.
Pharmacol Biochem Behav ; 211: 173290, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34662589

RESUMO

Alcohol abuse is a widespread cause of aggressive and impulsive behaviors that impact the users as well as their entourage. However, only a few medications are effective. Recently, cannabidiol has been reported to improve mood disorders and recovery from substance abuse, yet the psychopharmacologic effects of cannabidiol in ethanol-induced drug reward and aggressivity remain unexplored. In the present study, we investigated the effects of cannabidiol on ethanol-induced place preference and aggressivity in individually and group-housed male rats using the conditioned place preference test, and intruder evoc aggression test, respectively. The obtained results showed that ethanol significantly increased locomotor activity, induced conditioned place preference in all animals, and, specifically, increased aggressivity in individually housed rats. These behavioural impairments induced by ethanol were associated with decreased glucocorticoid and mineralocorticoid receptors transcription in the prefrontal cortex. Notwithstanding, cannabidiol at a dose of 10 mg/kg significantly inhibited Et-OH-induced place preference in group-housed, but not in individually housed rats, and markedly inhibited the aggressive behaviour. These findings suggest that ethanol-induced behavioural impairments are dependent on the housing condition that may affect corticosterone receptors expression and subsequently the animal responsivity to cannabidiol treatment.


Assuntos
Agressão/efeitos dos fármacos , Canabidiol/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Etanol/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Abrigo para Animais , Locomoção/efeitos dos fármacos , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar , Recompensa
13.
Neurobiol Learn Mem ; 185: 107526, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34562619

RESUMO

Heightened fear responding is characteristic of fear- and anxiety-related disorders, including post-traumatic stress disorder. Neural plasticity in the amygdala is essential for both initial fear learning and fear expression, and strengthening of synaptic connections between the medial geniculate nucleus (MgN) and amygdala is critical for auditory fear learning. However, very little is known about what happens in the MgN-amygdala pathway during fear recall and extinction, in which conditional fear decreases with repeated presentations of the auditory stimulus alone. In the present study, we found that optogenetic inhibition of activity in the MgN-amygdala pathway during fear retrieval and extinction reduced expression of conditional fear. While this effect persisted for at least two weeks following pathway inhibition, it was specific to the context in which optogenetic inhibition occurred, linking MgN-BLA inhibition to facilitation of extinction-like processes. Reduced fear expression through inhibition of the MgN-amygdala pathway was further characterized by similar synaptic expression of GluA1 and GluA2 AMPA receptor subunits compared to what was seen in controls. Inhibition also decreased CREB phosphorylation in the amygdala, similar to what has been reported following auditory fear extinction. We then demonstrated that this effect was reduced by inhibition of GluN2B-containing NMDA receptors. These results demonstrate a new and important role for the MgN-amygdala pathway in extinction-like processes, and show that suppressing activity in this pathway results in a persistent decrease in fear behavior.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Corpos Geniculados/fisiologia , Vias Neurais/fisiologia , Estimulação Acústica , Animais , Condicionamento Clássico/efeitos dos fármacos , Extinção Psicológica/fisiologia , Imunofluorescência , Hylobatidae , Masculino , Optogenética , Piperidinas/farmacologia , Ratos , Ratos Long-Evans , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia
14.
Neurosci Lett ; 762: 136156, 2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34358624

RESUMO

Pleiotrophin (PTN) and midkine (MK) are growth factors that modulate alcohol consumption and reward. Since both PTN and MK limit the rewarding effects of alcohol, pharmacological potentiation of the PTN and MK signaling pathways has been proposed for the treatment of alcohol use disorders (AUD). Although the use of this therapy in the prevention of alcohol relapse is important, the potential role of these cytokines in extinguishing alcohol-induced seeking behavior is a key question that remains unanswered. To fill this gap, we have now studied the extinction of the conditioned place preference (CPP) induced by different doses of alcohol in Ptn knockout (Ptn-/-) and Mk knockout (Mk-/-) mice. The data confirm a higher sensitivity of Ptn-/- mice to the conditioning effects of a low dose (1 g/kg) and a rewarding dose (2 g/kg) of alcohol, while Mk-/- mice are only more susceptible to the conditioning effects of the low dose of this drug. More importantly, the percentage of Mk-/- mice, not Ptn-/- mice, that efficiently extinguished alcohol-induced CPP was significantly higher than that of Wt mice. Taken together, the data presented here confirm that Ptn and Mk are genetic factors that determine the conditioning effects of alcohol in mice and that Mk is a novel factor that plays an important role in the extinction of alcohol-induced CPP.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Alcoolismo/genética , Condicionamento Clássico/efeitos dos fármacos , Extinção Psicológica/fisiologia , Midkina/metabolismo , Animais , Proteínas de Transporte/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Citocinas/metabolismo , Comportamento de Procura de Droga/fisiologia , Etanol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Behav Brain Res ; 415: 113515, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34371088

RESUMO

The neuropeptide orexin-A (OX-A) has diverse functions, including maintaining arousal, autonomic control, motor activity and stress responses. These functions are regulated at different terminal regions where OX-A is released. The current study examined the physiological and behavioural effects of OX-A microinjections into the central amygdala (CeA) under basal and stressed conditions in rats. When OX-A was microinjected into the CeA and the animals returned to the home-cage, heart rate and mean arterial pressure were increased compared to vehicle-injected controls. General activity of the animal was also increased, indicating that OX-A activity in CeA contributes to increased arousal. This outcome is similar to the effects of central intracerebroventricular infusions of OX-A, as well as the cardiovascular effects previously demonstrated at many of OX's efferent hypothalamic and brainstem structures. In a second study, animals were fear-conditioned to a context by delivery of electric footshocks and then animals were re-exposed to the conditioned context at test. When OX-A was microinjected at test, freezing behaviour was reduced and there was a corresponding increase in the animal's activity but no impact on the pressor and cardiac responses (i.e, blood pressure and heart rate were unchanged). This reduction in freezing suggests that OX-A activates amygdala neurons that inhibit freezing, which is similar to the actions of other neuropeptides in the CeA that modulate the appropriate defence response to fearful stimuli. Overall, these data indicate that the CeA is an important site of OX-A modulation of cardiovascular and motor activity, as well as conditioned freezing responses.


Assuntos
Comportamento Animal/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Núcleo Central da Amígdala/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Medo/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Orexinas/farmacologia , Animais , Masculino , Orexinas/administração & dosagem , Ratos , Ratos Wistar
16.
Neurosci Lett ; 762: 136164, 2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34371124

RESUMO

Cocaine use disorder is a serious problem worldwide, and there are no approved medications for its treatment. A novel approach to the treatment of drug addiction is the use of natural products, and, in this context, preclinical evidence suggests that Hypericum perforatum L. (Hypericum) is effective against alcohol and other substance use disorders. We hypothesised that Hypericum could also be useful as a treatment for cocaine use disorder, and so we set out to test its effectiveness in a mice model of cocaine addiction. In the first experiment we evaluated its effects on the acquisition of cocaine-induced conditioned place preference (CPP). Adult male mice were conditioned with cocaine (25 mg/kg), cocaine with Hypericum (75, 150 or 300 mg/kg) or the plant extract alone (300 mg/kg). In the second experiment, we tested the effects of Hypericum on stress-induced reinstatement of cocaine CPP. All the mice were conditioned with cocaine (25 mg/kg) and, after extinction of CPP, the reinstating effects of social defeat (alone or with 75, 150 or 300 mg/kg of Hypericum) were evaluated. All the doses of Hypericum prevented the acquisition of cocaine-induced CPP. Furthermore, the plant extract dose-dependently reduced the reinstating effects of social defeat. Therefore, Hypericum is effective in reducing the rewarding effects of cocaine and prevents the stress-induced reinstatement of cocaine CPP in mice. The mechanisms underlying these positive effects of Hypericum perforatum L. need to be determined by future research. Our results endorse Hypericum as a natural treatment for cocaine dependence.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Condicionamento Clássico/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Hypericum , Extratos Vegetais/farmacologia , Animais , Masculino , Camundongos
17.
Int J Mol Sci ; 22(15)2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34361071

RESUMO

3,4-Methylenedioxypyrovalerone (MDPV) is a new psychoactive substance (NPS) and the most widespread and life-threatening synthetic cathinone of the "bath salts". Preclinical research has proven the cocaine-like psychostimulant effects of MDPV and its potential for abuse. Cannabidiol (CBD) is a non-psychotropic phytocannabinoid that has emerged as a new potential treatment for drug addiction. Here, we tested the effects of CBD (20 mg/kg) on MDPV (2 mg/kg)-induced conditioned place preference and MDPV (0.05 and 0.075 mg/kg/infusion) self-administration paradigms. In addition, we assessed the effects of the co-administration of CBD and MDPV (3 and 4 mg/kg) on anxiety-like behaviour using the elevated plus maze (EPM). CBD mitigated the MDPV-induced conditioned place preference. On the contrary, CBD administration throughout the MDPV (0.075 mg/kg/infusion) self-administration increased drug-seeking and taking behaviours, but only in the high-responders group of mice. Furthermore, CBD exerted anxiolytic-like effects, exclusively in MDPV-treated mice. Taken together, our results indicate that CBD modulation of MDPV-induced motivational responses in mice varies depending on the requirements of the learning task, resulting in a complex response. Therefore, further research attempting to decipher the behavioural and molecular interactions between CBD and MDPV is needed.


Assuntos
Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Benzodioxóis/toxicidade , Canabidiol/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Pirrolidinas/toxicidade , Inibidores da Captação Adrenérgica/toxicidade , Animais , Anticonvulsivantes/farmacologia , Ansiedade/induzido quimicamente , Ansiedade/patologia , Condicionamento Clássico/efeitos dos fármacos , Masculino , Camundongos , Catinona Sintética
18.
Behav Brain Res ; 415: 113521, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34391796

RESUMO

Methamphetamine withdrawal can induce intense cravings leading to relapse. Contexts/cues paired with chronic methamphetamine use develop incentive motivational properties, promoting future drug-seeking and taking behavior. Research has shown that, in adult male rats, the selective 5-HT2A receptor antagonist M100907 attenuates the acquisition of methamphetamine-induced conditioned place preference (CPP), a measure that examines conditioned associations between the rewarding properties of drugs and contexts. However, these findings have not been extended to adult female rats. The present study investigated the effects of M100907 on the acquisition of methamphetamine-CPP in adult female rats. During conditioning, rats were administered M100907 (0, 0.025, 0.25 mg/kg, i.p.) 15 min before methamphetamine (1 mg/kg, i.p.) and then placed into their initially non-preferred chamber for 30 min, or administered saline and placed into their initially preferred chamber for 30 min. Conditioning sessions were separated by four hours. Following four days of conditioning, the effects of M100907 on the acquisition of methamphetamine-CPP were assessed during a 15 min drug-free test trial. Pretreatment with M100907 dose-dependently attenuated the acquisition of methamphetamine-induced CPP. Blocking 5-HT2A receptors with a low dose of the selective antagonist M100907 attenuated the rewarding effects of methamphetamine in adult female rats. These data provide further evidence that the 5-HT2A receptor subtype is involved in the behavioral effects of methamphetamine.


Assuntos
Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Metanfetamina/farmacologia , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Feminino , Fluorbenzenos/farmacologia , Masculino , Metanfetamina/administração & dosagem , Piperidinas/farmacologia , Ratos , Ratos Long-Evans , Antagonistas do Receptor 5-HT2 de Serotonina/administração & dosagem
19.
Behav Brain Res ; 415: 113517, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34389427

RESUMO

Cannabis use disorder (CUD) has doubled in prevalence over the past decade as a nation-wide trend toward legalization allows for increased drug accessibility. As a result, marijuana has become the most commonly used illicit drug in the United States particularly among the adolescent population. This is especially concerning since there is greater risk for the harmful side effects of drug use during this developmental period due to ongoing brain maturation. Increasing evidence indicates that CUD often occurs along with other debilitating conditions including both alcohol use disorder (AUD) and anxiety disorders such post-traumatic stress disorder (PTSD). Additionally, exposure to cannabis, alcohol, and stress can induce alterations in glutamate regulation and homeostasis in the prefrontal cortex (PFC) that may lead to impairments in neuronal functioning and cognition. Therefore, in order to study the relationship between drug exposure and the development of PTSD, these studies utilized rodent models to determine the impact of adolescent exposure to delta-9-tetrahydrocannabinol (THC) and ethanol on responses to fear stimuli during fear conditioning and used calcium imaging to measure glutamate activity in the prelimbic cortex during this behavioral paradigm. The results from these experiments indicate that adolescent exposure to THC and ethanol leads to enhanced sensitivity to fear stimuli both behaviorally and neuronally. Additionally, these effects were attenuated when animals were treated with the glutamatergic modulator N-acetylcysteine (NAC). In summary, these studies support the hypothesis that adolescent exposure to THC and ethanol leads to alterations in fear stimuli processing through glutamatergic reliant modifications in PFC signaling.


Assuntos
Comportamento Animal/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Dronabinol/farmacologia , Etanol/farmacologia , Medo/efeitos dos fármacos , Fatores Etários , Animais , Agonistas de Receptores de Canabinoides/administração & dosagem , Depressores do Sistema Nervoso Central/administração & dosagem , Dronabinol/administração & dosagem , Etanol/administração & dosagem , Masculino , Ratos , Ratos Wistar
20.
Neuropharmacology ; 196: 108717, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34273388

RESUMO

Drugs of abuse, including morphine and cocaine, can reduce hippocampal neurogenesis (HN). Whereas promotion of HN is being increasingly recognized as a promising strategy for treating morphine and cocaine addiction. The present study is focused on exploring the changes of HN during methamphetamine (METH) administration and further clarify if HN is involved in METH-associated reward memory. After successfully establishing the conditioned place preference (CPP) paradigm to simulate the METH-associated reward memory in C57BL/6 mice, we observed that HN was significantly inhibited during METH (2 mg/kg, i. p.) administration and returned to normal after the extinction of METH CPP, as indicated by the immunostaining of bromodeoxyuridine (BrdU) and doublecortin (DCX) in the hippocampus. To promote/inhibit HN levels, 7,8-dihydroxyflavone (DHF), a small tyrosine kinase receptor B (TrkB) agonist and temozolomide (TMZ), an alkylating agent, were administered intraperitoneally (i.p.), respectively. The data showed that either DHF (5 mg/kg, i. p.) or TMZ (25 mg/kg, i. p.) pre-treatment before METH administration could significantly prolong extinction and enhance reinstatement of the reward memory. Notably, DHF treatment after METH administration significantly facilitated extinction and inhibited METH reinstatement, while TMZ treatment resulted in opposite effects. The present study indicated that METH administration could induce a temporal inhibitory effect on HN. More importantly, promotion of HN after the acquisition of METH-associated reward memory, but not inhibition of HN or promotion of HN before the acquisition of reward memory, could facilitate METH extinction and inhibit METH reinstatement, indicating the beneficial effect of HN on METH addiction by erasing the according reward memory.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Metanfetamina/farmacologia , Neurogênese/efeitos dos fármacos , Recompensa , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Extinção Psicológica/efeitos dos fármacos , Flavonas/farmacologia , Hipocampo/metabolismo , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Temozolomida/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...