Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.277
Filtrar
1.
Sci Adv ; 10(17): eadl6554, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38657057

RESUMO

MDMA (3,4-methylenedioxymethamphetamine) is a psychoactive drug with powerful prosocial effects. While MDMA is sometimes termed an "empathogen," empirical studies have struggled to clearly demonstrate these effects or pinpoint underlying mechanisms. Here, we paired the social transfer of pain and analgesia-behavioral tests modeling empathy in mice-with region-specific neuropharmacology, optogenetics, and transgenic manipulations to explore MDMA's action as an empathogen. We report that MDMA, given intraperitoneally or infused directly into the nucleus accumbens (NAc), robustly enhances the social transfer of pain and analgesia. Optogenetic stimulation of 5-HT release in the NAc recapitulates the effects of MDMA, implicating 5-HT signaling as a core mechanism. Last, we demonstrate that systemic MDMA or optogenetic stimulation of NAc 5-HT inputs restores deficits in empathy-like behaviors in the Shank3-deficient mouse model of autism. These findings demonstrate enhancement of empathy-related behaviors by MDMA and implicate 5-HT signaling in the NAc as a core mechanism mediating MDMA's empathogenic effects.


Assuntos
Empatia , Proteínas dos Microfilamentos , N-Metil-3,4-Metilenodioxianfetamina , Núcleo Accumbens , Optogenética , Serotonina , Animais , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Empatia/efeitos dos fármacos , Serotonina/metabolismo , Camundongos , Masculino , Comportamento Animal/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Transtorno Autístico/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
2.
Behav Neurosci ; 138(2): 108-124, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38661670

RESUMO

The cannabinoid system is being researched as a potential pharmaceutical target for a multitude of disorders. The present study examined the effect of indirect and direct cannabinoid agonists on mesolimbic dopamine release and related behaviors in C57BL/6J (B6) mice. The indirect cannabinoid agonist N-arachidonoyl serotonin (AA-5-HT) indirectly agonizes the cannabinoid system by preventing the metabolism of endocannabinoids through fatty acid amide hydrolase inhibition while also inhibiting transient receptor potential vanilloid Type 1 channels. Effects of AA-5-HT were compared with the direct cannabinoid receptor Type 1 agonist arachidonoyl-2'-chloroethylamide (ACEA). In Experiment 1, mice were pretreated with seven daily injections of AA-5-HT, ACEA, or vehicle prior to assessments of locomotor activity using open field (OF) testing and phasic dopamine release using in vivo fixed potential amperometry. Chronic exposure to AA-5-HT did not alter locomotor activity or mesolimbic dopamine functioning. Chronic exposure to ACEA decreased rearing and decreased phasic dopamine release while increasing the dopaminergic response to cocaine. In Experiment 2, mice underwent AA-5-HT, ACEA, or vehicle conditioned place preference, then saccharin preference testing, a measure commonly associated with anhedonia. Mice did not develop a conditioned place preference or aversion for AA-5-HT or ACEA, and repeated exposure to AA-5-HT or ACEA did not alter saccharin preference. Altogether, the findings suggest that neither of these drugs induce behaviors that are classically associated with abuse liability in mice; however, direct cannabinoid receptor Type 1 agonism may play more of a role in mediating mesolimbic dopamine functioning than indirect cannabinoid agonism. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Assuntos
Agonistas de Receptores de Canabinoides , Dopamina , Camundongos Endogâmicos C57BL , Animais , Dopamina/metabolismo , Masculino , Camundongos , Agonistas de Receptores de Canabinoides/farmacologia , Serotonina/metabolismo , Locomoção/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Ácidos Araquidônicos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Cocaína/farmacologia , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Atividade Motora/efeitos dos fármacos
3.
Arch Pharm Res ; 47(4): 360-376, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38551761

RESUMO

Novel psychoactive substances (NPSs) are new psychotropic drugs designed to evade substance regulatory policies. 25E-NBOMe (2-(4-ethyl-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine) has recently been identified as an NPS, and its recreational misuse has been reported to be rapidly increasing. However, the psychopharmacological effects and mechanisms of 25E-NBOMe have not been studied. We examined the abuse potential of 25E-NBOMe using the conditioned place preference in male mice and self-administration paradigms in male rats. Additionally, immunoblot assay, enzyme-linked immunosorbent assay, and microdialysis were used to determine the molecular effects of 25E-NBOMe in the nucleus accumbens (NAc). Our data demonstrated that 25E-NBOMe induces conditioned place preference, and the dopaminergic signaling in the NAc mediates these. Following 25E-NBOMe administration, expression of dopamine transporter and dopamine D1 receptor (D1DR) were enhanced in the NAc of male mice, and NAc dopamine levels were reduced in both male mice and rats. Induction of intracellular dopaminergic pathways, DARPP32, and phosphorylation of CREB in the NAc of male mice was also observed. Significantly, pharmacological blockade of D1DR or chemogenetic inhibition of D1DR-expressing medium spiny neurons in the NAc attenuated 25E-NBOMe-induced conditioned place preference in male mice. We also examined the hallucinogenic properties of 25E-NBOMe using the head twitch response test in male mice and found that this behavior was mediated by serotonin 2A receptor activity. Our findings demonstrate that D1DR signaling may govern the addictive potential of 25E-NBOMe. Moreover, our study provides new insights into the potential mechanisms of substance use disorder and the improvement of controlled substance management.


Assuntos
Núcleo Accumbens , Psicotrópicos , Receptores de Dopamina D1 , Recompensa , Transdução de Sinais , Animais , Masculino , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/agonistas , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ratos , Psicotrópicos/farmacologia , Ratos Sprague-Dawley , Camundongos Endogâmicos C57BL , Fenetilaminas/farmacologia , Autoadministração , Dopamina/metabolismo
4.
Nature ; 618(7966): 790-798, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37316665

RESUMO

Psychedelics are a broad class of drugs defined by their ability to induce an altered state of consciousness1,2. These drugs have been used for millennia in both spiritual and medicinal contexts, and a number of recent clinical successes have spurred a renewed interest in developing psychedelic therapies3-9. Nevertheless, a unifying mechanism that can account for these shared phenomenological and therapeutic properties remains unknown. Here we demonstrate in mice that the ability to reopen the social reward learning critical period is a shared property across psychedelic drugs. Notably, the time course of critical period reopening is proportional to the duration of acute subjective effects reported in humans. Furthermore, the ability to reinstate social reward learning in adulthood is paralleled by metaplastic restoration of oxytocin-mediated long-term depression in the nucleus accumbens. Finally, identification of differentially expressed genes in the 'open state' versus the 'closed state' provides evidence that reorganization of the extracellular matrix is a common downstream mechanism underlying psychedelic drug-mediated critical period reopening. Together these results have important implications for the implementation of psychedelics in clinical practice, as well as the design of novel compounds for the treatment of neuropsychiatric disease.


Assuntos
Período Crítico Psicológico , Alucinógenos , Aprendizagem , Recompensa , Animais , Humanos , Camundongos , Estado de Consciência/efeitos dos fármacos , Alucinógenos/farmacologia , Alucinógenos/uso terapêutico , Aprendizagem/efeitos dos fármacos , Fatores de Tempo , Ocitocina/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos
5.
Behav Brain Res ; 437: 114127, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36174843

RESUMO

The 5-hydroxytryptamine 2A (5-HT2A) receptor plays an important role in schizophrenia. The 5-HT2A receptor is also involved in the regulation of prepulse inhibition (PPI) in rodents. The aim of this study was to determine whether selective 5-HT2A receptor agonizts or antagonists may alter PPI in rats and to identify the critical brain regions in which the activity of 5-HT2A receptors regulates PPI. The results showed that infusion of the 5-HT2A receptor agonist TCB-2 into the lateral ventricle disrupted PPI, but the 5-HT2A receptor antagonist M100907 had no such effect. In addition, local infusion of TCB-2 into the nucleus accumbens and ventral pallidum disrupted PPI, whereas the same manipulation in the medial prefrontal cortex, ventral hippocampus, and ventral tegmental area did not disrupt PPI. In conclusion, agonism of 5-HT2A receptors in the ventral pallidum and nucleus accumbens can disrupt PPI. The ventral pallidum and nucleus accumbens are critical brain regions responsible for the regulation of PPI by serotonin. These findings contribute to the extensive exploration of the molecular and neural mechanisms underlying the regulatory effect of 5-HT2A receptor activity on PPI, especially the neural circuits modulated by 5-HT2A receptor activity.


Assuntos
Prosencéfalo Basal , Núcleo Accumbens , Inibição Pré-Pulso , Receptor 5-HT2A de Serotonina , Agonistas do Receptor 5-HT2 de Serotonina , Animais , Ratos , Prosencéfalo Basal/efeitos dos fármacos , Prosencéfalo Basal/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Inibição Pré-Pulso/efeitos dos fármacos , Ratos Sprague-Dawley , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia
6.
Nature ; 608(7922): 368-373, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35896744

RESUMO

Ketamine is used clinically as an anaesthetic and a fast-acting antidepressant, and recreationally for its dissociative properties, raising concerns of addiction as a possible side effect. Addictive drugs such as cocaine increase the levels of dopamine in the nucleus accumbens. This facilitates synaptic plasticity in the mesolimbic system, which causes behavioural adaptations and eventually drives the transition to compulsion1-4. The addiction liability of ketamine is a matter of much debate, in part because of its complex pharmacology that among several targets includes N-methyl-D-aspartic acid (NMDA) receptor (NMDAR) antagonism5,6. Here we show that ketamine does not induce the synaptic plasticity that is typically observed with addictive drugs in mice, despite eliciting robust dopamine transients in the nucleus accumbens. Ketamine nevertheless supported reinforcement through the disinhibition of dopamine neurons in the ventral tegmental area (VTA). This effect was mediated by NMDAR antagonism in GABA (γ-aminobutyric acid) neurons of the VTA, but was quickly terminated by type-2 dopamine receptors on dopamine neurons. The rapid off-kinetics of the dopamine transients along with the NMDAR antagonism precluded the induction of synaptic plasticity in the VTA and the nucleus accumbens, and did not elicit locomotor sensitization or uncontrolled self-administration. In summary, the dual action of ketamine leads to a unique constellation of dopamine-driven positive reinforcement, but low addiction liability.


Assuntos
Ketamina , Transtornos Relacionados ao Uso de Substâncias , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Ketamina/efeitos adversos , Ketamina/farmacologia , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Reforço Psicológico , Autoadministração , Transtornos Relacionados ao Uso de Substâncias/etiologia , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos
7.
Brain Res Bull ; 181: 157-166, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35122898

RESUMO

Pramipexole is a potent agonist of D3 and D2 dopamine receptors, currently approved for clinical use in Parkinson's disease (PD) and restless leg syndrome. Several studies have shown that pramipexole significantly increases the risk of pathological gambling and impulse-control disorders. While these iatrogenic complications can impose a severe social and financial burden, their treatment poses serious clinical challenges. Our group previously reported that the steroidogenic inhibitor finasteride reduced pathological gambling severity in PD patients who developed this complication following pramipexole treatment. To study the mechanisms underlying these effects, here we tested the impact of finasteride in a rat model of pramipexole-induced alterations of probability discounting. We previously showed that, in rats exposed to low doses of the monoamine-depleting agent reserpine (1 mg/kg/day, SC), pramipexole (0.3 mg/kg/day, SC) increased the propensity to engage in disadvantageous choices. This effect was paralleled by a marked D3 receptor upregulation in the nucleus accumbens. First, we tested how finasteride (25-50 mg/kg, IP) intrinsically affects probability discounting. While the highest dose of finasteride produced a marked lack of interest in lever pressing (manifested as a significant increase in omissions), the 25 mg/kg (IP) dose did not intrinsically modify probability discounting. However, this finasteride regimen significantly reduced the adverse effects of reserpine and pramipexole in probability discounting by diminishing rats' propensity to engage in highly disadvantageous probabilistic choices. The same regimen also reversed the upregulation of D3 receptors in the nucleus accumbens induced by reserpine and pramipexole. These findings confirm that finasteride opposes the impulsivity caused by pramipexole and suggest that this effect may be underpinned by a normalizing effect on D3 receptor expression in the nucleus accumbens.


Assuntos
Inibidores de 5-alfa Redutase/farmacologia , Comportamento de Escolha/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Finasterida/farmacologia , Comportamento Impulsivo/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Pramipexol/farmacologia , Aprendizagem por Probabilidade , Receptores de Dopamina D3/efeitos dos fármacos , Receptores de Dopamina D3/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Ratos , Receptores de Dopamina D3/agonistas
8.
Artigo em Inglês | MEDLINE | ID: mdl-34973413

RESUMO

Gender is considered as a pivotal determinant of mental health. Indeed, several psychiatric disorders such as anxiety and depression are more common and persistent in women than in men. In the past two decades, impaired brain energy metabolism has been highlighted as a risk factor for the development of these psychiatric disorders. However, comprehensive behavioural and neurobiological studies in brain regions relevant to anxiety and depression symptomatology are scarce. In the present study, we summarize findings describing cannabidiol effects on anxiety and depression in maternally separated female mice as a well-established rodent model of early-life stress associated with many mental disorders. Our results indicate that cannabidiol could prevent anxiolytic- and depressive-related behaviour in early-life stressed female mice. Additionally, maternal separation with early weaning (MSEW) caused long-term changes in brain oxidative metabolism in both nucleus accumbens and amygdalar complex measured by cytochrome c oxidase quantitative histochemistry. However, cannabidiol treatment could not revert brain oxidative metabolism impairment. Moreover, we identified hyperphosphorylation of mTOR and ERK 1/2 proteins in the amygdala but not in the striatum, that could also reflect altered brain intracellular signalling related with to bioenergetic impairment. Altogether, our study supports the hypothesis that MSEW induces profound long-lasting molecular changes in mTOR signalling and brain energy metabolism related to depressive-like and anxiety-like behaviours in female mice, which were partially ameliorated by CBD administration.


Assuntos
Anticonvulsivantes/administração & dosagem , Ansiedade/tratamento farmacológico , Canabidiol/administração & dosagem , Emoções/fisiologia , Privação Materna , Núcleo Accumbens/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Comportamento Animal , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Camundongos , Serina-Treonina Quinases TOR/genética , Desmame
9.
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
10.
Int J Neuropsychopharmacol ; 25(5): 425-431, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35022720

RESUMO

Schizophrenia is a severe mental disorder featuring psychotic, depressive, and cognitive alterations. Current antipsychotic drugs preferentially target dopamine D2-R and/or serotonergic 5-HT2A/1A-R. They partly alleviate psychotic symptoms but fail to treat negative symptoms and cognitive deficits. Here we report on the putative antipsychotic activity of (1-[(3-fluorophenyl)sulfonyl]-4-(piperazin-1-yl)-1H-pyrrolo[3,2-c]quinoline dihydrochloride) (FPPQ), a dual serotonin 5-HT3-R/5-HT6-R antagonist endowed with pro-cognitive properties. FPPQ fully reversed phencyclidine-induced decrease of low-frequency oscillations in the medial prefrontal cortex of anaesthetized rats, a fingerprint of antipsychotic activity. This effect was mimicked by the combined administration of the 5-HT3-R and 5-HT6-R antagonists ondansetron and SB-399 885, respectively, but not by either drug alone. In freely moving rats, FPPQ countered phencyclidine-induced hyperlocomotion and augmentation of gamma and high-frequency oscillations in medial prefrontal cortex, dorsal hippocampus, and nucleus accumbens. Overall, this supports that simultaneous blockade of 5-HT3R and 5-HT6-R-like that induced by FPPQ-can be a new target in antipsychotic drug development.


Assuntos
Antipsicóticos , Encéfalo , Fenciclidina , Quinolinas , Antagonistas da Serotonina , Animais , Antipsicóticos/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Fenciclidina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Quinolinas/farmacologia , Ratos , Receptores de Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia
11.
J Neurosci ; 42(9): 1738-1751, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35042768

RESUMO

Striatal adenosine A1 receptor (A1R) activation can inhibit dopamine release. A1Rs on other striatal neurons are activated by an adenosine tone that is limited by equilibrative nucleoside transporter 1 (ENT1) that is enriched on astrocytes and is ethanol sensitive. We explored whether dopamine release in nucleus accumbens core is under tonic inhibition by A1Rs, and is regulated by astrocytic ENT1 and ethanol. In ex vivo striatal slices from male and female mice, A1R agonists inhibited dopamine release evoked electrically or optogenetically and detected using fast-scan cyclic voltammetry, most strongly for lower stimulation frequencies and pulse numbers, thereby enhancing the activity-dependent contrast of dopamine release. Conversely, A1R antagonists reduced activity-dependent contrast but enhanced evoked dopamine release levels, even for single optogenetic pulses indicating an underlying tonic inhibition. The ENT1 inhibitor nitrobenzylthioinosine reduced dopamine release and promoted A1R-mediated inhibition, and, conversely, virally mediated astrocytic overexpression of ENT1 enhanced dopamine release and relieved A1R-mediated inhibition. By imaging the genetically encoded fluorescent adenosine sensor [GPCR-activation based (GRAB)-Ado], we identified a striatal extracellular adenosine tone that was elevated by the ENT1 inhibitor and sensitive to gliotoxin fluorocitrate. Finally, we identified that ethanol (50 mm) promoted A1R-mediated inhibition of dopamine release, through diminishing adenosine uptake via ENT1. Together, these data reveal that dopamine output dynamics are gated by a striatal adenosine tone, limiting amplitude but promoting contrast, regulated by ENT1, and promoted by ethanol. These data add to the diverse mechanisms through which ethanol modulates striatal dopamine, and to emerging datasets supporting astrocytic transporters as important regulators of striatal function.SIGNIFICANCE STATEMENT Dopamine axons in the mammalian striatum are emerging as strategic sites where neuromodulators can powerfully influence dopamine output in health and disease. We found that ambient levels of the neuromodulator adenosine tonically inhibit dopamine release in nucleus accumbens core via adenosine A1 receptors (A1Rs), to a variable level that promotes the contrast in dopamine signals released by different frequencies of activity. We reveal that the equilibrative nucleoside transporter 1 (ENT1) on astrocytes limits this tonic inhibition, and that ethanol promotes it by diminishing adenosine uptake via ENT1. These findings support the hypotheses that A1Rs on dopamine axons inhibit dopamine release and, furthermore, that astrocytes perform important roles in setting the level of striatal dopamine output, in health and disease.


Assuntos
Astrócitos , Dopamina , Transportador Equilibrativo 1 de Nucleosídeo , Etanol , Núcleo Accumbens , Receptor A1 de Adenosina , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Dopamina/metabolismo , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Etanol/farmacologia , Feminino , Masculino , Camundongos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptor A1 de Adenosina/metabolismo
12.
Biochem Biophys Res Commun ; 588: 23-28, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34942530

RESUMO

Morphine is the pain releasing and abusing drug. Morphine leads to addiction by activating dopaminergic rewarding system consisted of the ventral tegmental area (VTA) and nucleus accumbens (NAc). Cholecystokinin (CCK) is a gut-brain neuropeptide and involved in morphine dependence. Brain-derived neurotrophic factor (BDNF) is a neurotrophin and plays roles in regulating addiction. Geranylgeranylacetone (GGA) is a medicine of protecting gastric mucosal injury and protecting neurons. Our previous study showed that GGA blocked morphine-induced withdrawal and relapse through inducing thioredoxin 1(Trx1). In this study, we investigated that whether cholecystokinin-B receptor (CCKB receptor) and BDNF were related to GGA inhibition on morphine addiction. At first, we made conditioned place preference (CPP) model and confirmed again that GGA blocked the expression of morphine-CPP in present study. Then, our results showed that morphine increased the expressions of dopamine D1 receptor, tyrosine hydroxylase (TH), CCKB receptor and BDNF in the VTA and NAc in mice, which was inhibited by GGA. These results suggest that CCK and BDNF in dopaminergic systems are associated with the role of GGA blocking morphine-CPP.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diterpenos/farmacologia , Morfina/efeitos adversos , Receptor de Colecistocinina B/metabolismo , Receptores de Dopamina D1/metabolismo , Animais , Condicionamento Clássico , Masculino , Camundongos Endogâmicos C57BL , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
13.
Neuropsychopharmacology ; 47(11): 1875-1882, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34916591

RESUMO

The nucleus accumbens shell (NAcSh) and its afferent and efferent neuronal projections control key aspects of motivation for cocaine. A recently described regulator of γ-aminobutyric acid (GABA) projections from the dorsal raphe nucleus (DRN) to the NAcSh (DRN → NAcSh) is the neuropeptide neuromedin U (NMU). Here, we find that systemic administration of NMU decreases breakpoint for cocaine on a progressive ratio schedule of reinforcement in male rats. Employing a retrograde adeno-associated virus (AAV), we found that RNAi-mediated knockdown of the NMU receptor 2 (NMUR2) in afferent DRN projections to the NAcSh increases the breakpoint for cocaine. Our previous studies demonstrated that NMU regulates GABA release in the NAcSh, and our current investigation found that systemic NMU administration suppresses cocaine-evoked GABA release in the NAcSh and increases phosphorylated c-Fos expression in neurons projecting from the NAcSh to the ventral pallidum (VP). To further probe the impact of NMU/NMUR2 on neuroanatomical pathways regulating motivation for cocaine, we employed multi-viral transsynaptic studies. Using a combination of rabies virus and retrograde AAV helper virus, we mapped the impact of NMU across three distinct brain regions simultaneously and found a direct connection of GABAergic DRN neurons to the NAcSh → VP pathway. Together, these data reveal that NMU/NMUR2 modulates a direct connection within the GABAergic DRN → NAcSh → VP circuit that diminishes breakpoints for cocaine. These findings importantly advance our understanding of the neurochemical underpinnings of pathway-specific regulation of neurocircuitry that may regulate cocaine self-administration, providing a unique therapeutic perspective.


Assuntos
Estimulantes do Sistema Nervoso Central , Cocaína , Neuropeptídeos , Núcleo Accumbens , Automedicação , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/administração & dosagem , Cocaína/farmacologia , Masculino , Motivação/efeitos dos fármacos , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ratos , Automedicação/psicologia , Ácido gama-Aminobutírico/metabolismo
14.
Behav Brain Res ; 418: 113660, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-34752844

RESUMO

Accumulating studies consistently show that methylphenidate (MPD), the first line drug for treating Attention-Deficit Hyperactivity Disorder (ADHD), is abused by patients to whom the drug is prescribed. Like other psychostimulants, only low doses of MPD improve cognitive performance while higher doses impair it. Preventing the use of high doses of MPD is important for retaining its therapeutic efficacy. Previously, it has been shown that performance in Morris water maze test is improved in rats treated, orally, with MPD in doses of 2.5 mg/kg; but higher doses (5 mg/kg) impair it. The present study is designed to monitor rewarding effects of 2.5 mg/kg MPD in conditioned place preference (CPP) paradigm and its potential inhibition in buspirone co-treated animals. Our results show that rewarding effects of MPD in CPP paradigm are prevented in rats co-treated with buspirone in doses of 0.1 and 0.3 mg/kg. Animals treated with MPD exhibit a downregulation of 5-HT1A receptor mRNA in the nucleus accumbens which is also prevented in rats co-treated with 0.1 and 0.3 mg/kg but not 1.0 and 2.0 mg/kg buspirone. Administration of buspirone in these doses is not rewarding in CPP test and upregulates 5-HT1A receptor mRNA in the nucleus accumbens. The findings suggest that co-use of low doses of buspirone can prevent rewarding effects of MPD to help retain its therapeutic efficacy.


Assuntos
Buspirona/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Metilfenidato/farmacologia , Núcleo Accumbens/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptor 5-HT1A de Serotonina , Agonistas do Receptor de Serotonina/farmacologia , Animais , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Relação Dose-Resposta a Droga , Masculino , Ratos , Receptor 5-HT1A de Serotonina/metabolismo , Recompensa
15.
Neuropharmacology ; 203: 108874, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34748860

RESUMO

RATIONALE: The nucleus accumbens (NAc) is important for regulating a number of behaviors, including alcohol and substance use. We previously found that chemogenetically manipulating neuronal activity in the NAc core regulates binge-like drinking in mice. The central amygdala (CeA) is also an important regulator of alcohol drinking, and projects to the NAc core. We tested whether neuronal projections from the CeA to the NAc core, or neuropeptides released by the CeA in the NAc core, could regulate binge drinking. METHODS: For experiment 1, mice were administered AAV2 Cre-GFP into the NAc core and a Cre-inducible DREADD [AAV2 DIO- hM3Dq, -hM4Di, or -mCherry control] into the CeA. We tested the effects of altering CeA to NAc core activity on binge-like ethanol intake (via "Drinking in the Dark", DID). For experiment 2, we bilaterally microinfused corticotropin releasing factor (CRF), neuropeptide Y (NPY), or somatostatin (SST) into the NAc core prior to DID. For experiment 3, we tested whether intra-NAc CRF antagonism prevented reductions in drinking induced by CNO/hM3Dq stimulation of CeA->NAc projections. RESULTS: Chemogenetically increasing activity in neurons projecting from the CeA to NAc core decreased binge-like ethanol drinking (p < 0.01). Intra-NAc core CRF mimicked chemogenetic stimulation of this pathway (p < 0.05). Binge-like drinking was unaffected by the doses of NPY and SST tested. Lastly, we found that intra-NAc CRF antagonism prevented reductions in drinking induced by chemogenetic stimulation of CeA->NAc projections. These findings demonstrate that neurons projecting from the CeA to NAc core that release CRF are capable of regulating binge-like drinking in mice.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Rede Nervosa/metabolismo , Núcleo Accumbens/metabolismo , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Hormônio Liberador da Corticotropina/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções/métodos , Rede Nervosa/efeitos dos fármacos , Neuropeptídeo Y/administração & dosagem , Núcleo Accumbens/efeitos dos fármacos , Piperazinas/administração & dosagem
16.
J Ethnopharmacol ; 284: 114824, 2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-34763040

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Mitragyna speciosa (Korth.) Havil., popularly known as Kratom (KT), is a medicinal plant used for pain suppression in Southeast Asia. It has been claimed to assist drug users withdraw from methamphetamine (METH) dependence. However, its use was controversial and not approved yet. AIM OF THE STUDY: This study was conducted to characterize local field potential (LFP) patterns in the nucleus accumbens (NAc) and the hippocampus (HP) in mice with METH conditioned place preference (CPP) that were treated with KT alkaloid extract. MATERIALS AND METHODS: Male Swiss albino ICR mice were implanted with intracraneal electrodes into the NAc and HP. To induce METH CPP, animals were injected intraperitoneally once a day with METH (1 mg/kg) and saline (0.9% w/v) alternately and put into METH/saline compartments to experience the associations between drug/saline injection and the unique environmental contexts for 10 sessions. Control group received saline injection paired with both saline/saline compartments. On post-conditioning day, effects of 40 (KT40), 80 (KT80) mg/kg KT alkaloid extract and 20 mg/kg bupropion (BP) on CPP scores and LFP powers and NAc-HP coherence were tested. RESULTS: Two-way ANOVA revealed significant induction of CPP by METH sessions (P < 0.01). Multiple comparisons indicated that METH CPP was completely abolished by KT80 (P < 0.001). NAc gamma I (30.0-44.9 Hz) and HP delta (1.0-3.9 Hz) powers were significantly increased in mice with METH CPP (P < 0.01). The elevated NAc gamma I was significantly suppressed by KT80 (P < 0.05) and the increased HP delta was significantly reversed by KT40 (P < 0.01) and KT80 (P < 0.001). In addition, NAc-HP coherence was also significantly increased in gamma I (30.0-44.9 Hz) frequency range (P < 0.05) but it was reversed by KT80 (P < 0.05). Treatment with BP did not produce significant effect on these parameters. CONCLUSIONS: These findings demonstrated that KT alkaloid extract significantly reversed CPP scores and LFP patterns induced by METH administration. The ameliorative effects of the extract might be beneficial for treatment of METH craving and addiction.


Assuntos
Alcaloides/farmacologia , Comportamento Aditivo/tratamento farmacológico , Metanfetamina/efeitos adversos , Mitragyna/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , Alcaloides/química , Animais , Comportamento Aditivo/induzido quimicamente , Estimulantes do Sistema Nervoso Central/efeitos adversos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Núcleo Accumbens/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/química
17.
Artigo em Inglês | MEDLINE | ID: mdl-34662693

RESUMO

Cannabidiol, as component of cannabis, can potentially hinder the rewarding impact of drug abuse; however, its mechanism is ambiguous. Moreover, the nucleus accumbens (NAc), as a key area in the reward circuit, extensively receives dopaminergic projections from the ventral tegmentum area. To elucidate the role of accumbal D1 and D2 dopamine receptor families in Cannabidiol's inhibitory impact on the acquisition and expression phases of methamphetamine (MET), the conditioned place preference (CPP) procedure as a common method to assay reward characteristics of drugs was carried out. Six groups of rats were treated by various doses of SCH23390 or Sulpiride (0.25, 1, and 4 µg/0.5 µL) in the NAc as D1 or D2 dopamine receptor family antagonists, respectively, prior to infusion of Cannabidiol (10 µg/5 µL) in the lateral ventricle (LV) over conditioning phase in the acquisition experiments. In the second step of the study, animals received SCH23390 or Sulpiride in the NAc before Cannabidiol (50 µg/5 µL) infusion into the LV in the expression phase of MET to illuminate the influence of SCH23390 or Sulpiride on the inhibitory impact of Cannabidiol on the expression of MET-induced CPP. Intra-NAc administration of either SCH23390 or Sulpiride impaired Cannabidiol's suppressive impact on the expression phase, while just Sulpiride could suppress the Cannabidiol's impact on the acquisition phase of the MET-induced CPP. Also, the inhibitory impact of Sulpiride was stranger in both phases of MET reward. It seems that Cannabidiol prevents the expression and acquisition phases of MET-induced CPP partly through the dopaminergic system in the NAc.


Assuntos
Canabidiol , Condicionamento Clássico , Metanfetamina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D2/efeitos dos fármacos , Recompensa , Animais , Benzazepinas/administração & dosagem , Canabidiol/administração & dosagem , Canabidiol/farmacologia , Antagonistas de Dopamina/administração & dosagem , Masculino , Ratos , Sulpirida/administração & dosagem , Área Tegmentar Ventral/efeitos dos fármacos
18.
Addict Biol ; 27(1): e13107, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34699111

RESUMO

Hazardous, heavy drinking increases risk for developing alcohol use disorder (AUD), which affects ~7% of adult Americans. Thus, understanding the molecular mechanisms promoting risk for heavy drinking is essential to developing more effective AUD pharmacotherapies than those currently approved by the FDA. Using genome-wide bisulfate sequencing, we identified DNA methylation (DNAm) signals within the nucleus accumbens core (NAcC) that differentiate nonheavy and heavy ethanol-drinking rhesus macaques. One differentially DNAm region (D-DMR) located within the gene neurobeachin (NBEA), which promotes synaptic membrane protein trafficking, was hypermethylated in heavy drinking macaques. A parallel study identified a similar NBEA D-DMR in human NAcC that distinguished alcoholic and nonalcoholic individuals. To investigate the role of NBEA in heavy ethanol drinking, we engineered a viral vector carrying a short hairpin RNA (shRNA) to reduce the expression of NBEA. Using two murine models of ethanol consumption: 4 days of drinking-in-the-dark and 4 weeks of chronic intermittent access, the knockdown of NBEA expression did not alter average ethanol consumption in either model. However, it did lead to a significant increase in the ethanol preference ratio. Following withdrawal, whole-cell patch clamp electrophysiological experiments revealed that Nbea knockdown led to an increase in spontaneous excitatory postsynaptic current amplitude with no alteration in spontaneous inhibitory postsynaptic currents, suggesting a specific role of NBEA in trafficking of glutamatergic receptors. Together, our findings suggest that NBEA could be targeted to modulate the preference for alcohol use.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Alcoolismo/genética , Proteínas de Transporte/genética , Proteínas do Tecido Nervoso/genética , Adulto , Idoso , Animais , Metilação de DNA/efeitos dos fármacos , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Núcleo Accumbens/efeitos dos fármacos
19.
Addict Biol ; 27(1): e13108, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34713509

RESUMO

Previous studies indicate that moderate-to-high ethanol (EtOH) concentrations enhance dopamine (DA) neurotransmission in the mesolimbic DA system from the ventral tegmental area (VTA) and projecting to the nucleus accumbens core (NAc). However, voltammetry studies demonstrate that moderate-to-high EtOH concentrations decrease evoked DA release at NAc terminals. The involvement of γ-aminobutyric acid (GABA) receptors (GABAA Rs), glycine (GLY) receptors (GLYRs) and cholinergic interneurons (CINs) in mediating EtOH inhibition of evoked NAc DA release were examined. Fast scan cyclic voltammetry, electrophysiology, optogenetics and immunohistochemistry techniques were used to evaluate the effects of acute and chronic EtOH exposure on DA release and CIN activity in C57/BL6, CD-1, transgenic mice and δ-subunit knockout (KO) mice (δ-/-). Ethanol decreased DA release in mice with an IC50 of 80 mM ex vivo and 2.0 g/kg in vivo. GABA and GLY decreased evoked DA release at 1-10 mM. Typical GABAA R agonists inhibited DA release at high concentrations. Typical GABAA R antagonists had minimal effects on EtOH inhibition of evoked DA release. However, EtOH inhibition of DA release was blocked by the α4 ß3 δ GABAA R antagonist Ro15-4513, the GLYR antagonist strychnine and by the GABA ρ1 (Rho-1) antagonist TPMPA (10 µM) and reduced significantly in GABAA R δ-/- mice. Rho-1 expression was observed in CINs. Ethanol inhibited GABAergic synaptic input to CINs from the VTA and enhanced firing rate, both of which were blocked by TPMPA. Results herein suggest that EtOH inhibition of DA release in the NAc is modulated by GLYRs and atypical GABAA Rs on CINs containing δ- and Rho-subunits.


Assuntos
Dopamina/metabolismo , Etanol/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de GABA/efeitos dos fármacos , Animais , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
20.
Acta Pharmacol Sin ; 43(2): 295-306, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34522005

RESUMO

Behavioral sensitization is a progressive increase in locomotor or stereotypic behaviours in response to drugs. It is believed to contribute to the reinforcing properties of drugs and to play an important role in relapse after cessation of drug abuse. However, the mechanism underlying this behaviour remains poorly understood. In this study, we showed that mTOR signaling was activated during the expression of behavioral sensitization to cocaine and that intraperitoneal or intra-nucleus accumbens (NAc) treatment with rapamycin, a specific mTOR inhibitor, attenuated cocaine-induced behavioural sensitization. Cocaine significantly modified brain lipid profiles in the NAc of cocaine-sensitized mice and markedly elevated the levels of phosphatidylinositol-4-monophosphates (PIPs), including PIP, PIP2, and PIP3. The behavioural effect of cocaine was attenuated by intra-NAc administration of LY294002, an AKT-specific inhibitor, suggesting that PIPs may contribute to mTOR activation in response to cocaine. An RNA-sequencing analysis of the downstream effectors of mTOR signalling revealed that cocaine significantly decreased the expression of SynDIG1, a known substrate of mTOR signalling, and decreased the surface expression of GluA2. In contrast, AAV-mediated SynDIG1 overexpression in NAc attenuated intracellular GluA2 internalization by promoting the SynDIG1-GluA2 interaction, thus maintaining GluA2 surface expression and repressing cocaine-induced behaviours. In conclusion, NAc SynDIG1 may play a negative regulatory role in cocaine-induced behavioural sensitization by regulating synaptic surface expression of GluA2.


Assuntos
Proteínas de Transporte/metabolismo , Cocaína/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de AMPA/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Biotinilação , Western Blotting , Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...