Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.649
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38731799

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder. Dopamine (DA) neurons in the substantia nigra pars compacta, which have axonal projections to the dorsal striatum (dSTR), degenerate in PD. In contrast, DA neurons in the ventral tegmental area, with axonal projections to the ventral striatum, including the nucleus accumbens (NAcc) shell, are largely spared. This study aims to uncover the relative contributions of glycolysis and oxidative phosphorylation (OxPhos) to DA release in the striatum. We measured evoked DA release in mouse striatal brain slices using fast-scan cyclic voltammetry applied every two minutes. Blocking OxPhos resulted in a greater reduction in evoked DA release in the dSTR when compared to the NAcc shell, while blocking glycolysis caused a more significant decrease in evoked DA release in the NAcc shell than in the dSTR. Furthermore, when glycolysis was bypassed in favor of direct OxPhos, evoked DA release in the NAcc shell decreased by approximately 50% over 40 min, whereas evoked DA release in the dSTR was largely unaffected. These results demonstrate that the dSTR relies primarily on OxPhos for energy production to maintain evoked DA release, whereas the NAcc shell depends more on glycolysis. Consistently, two-photon imaging revealed higher oxidation levels of DA terminals in the dSTR than in the NAcc shell. Together, these findings partly explain the selective vulnerability of DA terminals in the dSTR to degeneration in PD.


Assuntos
Corpo Estriado , Dopamina , Glicólise , Fosforilação Oxidativa , Animais , Dopamina/metabolismo , Camundongos , Corpo Estriado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Neurônios Dopaminérgicos/metabolismo , Núcleo Accumbens/metabolismo
2.
Elife ; 132024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38748470

RESUMO

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Assuntos
Neurônios Colinérgicos , Dopamina , Interneurônios , Optogenética , Dopamina/metabolismo , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/fisiologia , Ratos , Optogenética/métodos , Motivação , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Acetilcolina/metabolismo
3.
Addict Biol ; 29(5): e13397, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38711205

RESUMO

Neuronal ensembles in the medial prefrontal cortex mediate cocaine self-administration via projections to the nucleus accumbens. We have recently shown that neuronal ensembles in the prelimbic cortex form rapidly to mediate cocaine self-administration. However, the role of neuronal ensembles within the nucleus accumbens in initial cocaine-seeking behaviour remains unknown. Here, we sought to expand the current literature by testing the necessity of the cocaine self-administration ensemble in the nucleus accumbens core (NAcCore) 1 day after male and female rats acquire cocaine self-administration by using the Daun02 inactivation procedure. We found that disrupting the NAcCore ensembles after a no-cocaine reward-seeking test increased subsequent cocaine seeking, while disrupting NAcCore ensembles following a cocaine self-administration session decreased subsequent cocaine seeking. We then characterized neuronal cell type in the NAcCore using RNAscope in situ hybridization. In the no-cocaine session, we saw reduced dopamine D1 type neuronal activation, while in the cocaine self-administration session, we found preferential dopamine D1 type neuronal activity in the NAcCore.


Assuntos
Cocaína , Comportamento de Procura de Droga , Neurônios , Núcleo Accumbens , Autoadministração , Animais , Núcleo Accumbens/efeitos dos fármacos , Cocaína/farmacologia , Masculino , Feminino , Ratos , Comportamento de Procura de Droga/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Recompensa , Inibidores da Captação de Dopamina/farmacologia , Reforço Psicológico , Receptores de Dopamina D1 , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Ratos Sprague-Dawley , Córtex Pré-Frontal/efeitos dos fármacos
4.
CNS Neurosci Ther ; 30(5): e14737, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38702929

RESUMO

AIMS: This study aims to investigate the pharmacological effects and the underlying mechanism of cannabidiol (CBD) on methamphetamine (METH)-induced relapse and behavioral sensitization in male mice. METHODS: The conditioned place preference (CPP) test with a biased paradigm and open-field test were used to assess the effects of CBD on METH-induced relapse and behavioral sensitization in male mice. RNA sequencing and bioinformatics analysis was employed to identify differential expressed (DE) circRNAs, miRNAs, and mRNAs in the nucleus accumbens (NAc) of mice, and the interaction among them was predicted using competing endogenous RNAs (ceRNAs) network analysis. RESULTS: Chronic administration of CBD (40 mg/kg) during the METH withdrawal phase alleviated METH (2 mg/kg)-induced CPP reinstatement and behavioral sensitization in mice, as well as mood and cognitive impairments following behavioral sensitization. Furthermore, 42 DEcircRNAs, 11 DEmiRNAs, and 40 DEmRNAs were identified in the NAc of mice. The circMeis2-miR-183-5p-Kcnj5 network in the NAc of mice is involved in the effects of CBD on METH-induced CPP reinstatement and behavioral sensitization. CONCLUSIONS: This study constructed the ceRNAs network for the first time, revealing the potential mechanism of CBD in treating METH-induced CPP reinstatement and behavioral sensitization, thus advancing the application of CBD in METH use disorders.


Assuntos
Canabidiol , Metanfetamina , Camundongos Endogâmicos C57BL , MicroRNAs , RNA Circular , RNA Mensageiro , Animais , Canabidiol/farmacologia , Masculino , Metanfetamina/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Camundongos , RNA Circular/genética , RNA Mensageiro/metabolismo , Recidiva , Estimulantes do Sistema Nervoso Central/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Redes Reguladoras de Genes/efeitos dos fármacos
5.
Behav Pharmacol ; 35(4): 193-200, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38567425

RESUMO

Prepulse inhibition (PPI) is a crucial indicator of sensorimotor gating that is often impaired in neuropsychiatric diseases. Although dopamine D1 receptor agonists have been found to disrupt PPI in mice, the underlying mechanisms are not fully understood. In this study, we aimed to identify the brain regions responsible for the PPI-disruptive effect of the D1 agonist in mice. Results demonstrated that intraperitoneal administration of the selective dopamine D1 receptor agonist SKF82958 dramatically inhibited PPI, while the dopamine D1 receptor antagonist SCH23390 enhanced PPI. Additionally, local infusion of SKF82958 into the nucleus accumbens and medial prefrontal cortex disrupted PPI, but not in the ventral hippocampus. Infusion of SCH23390 into these brain regions also failed to enhance PPI. Overall, the study suggests that the nucleus accumbens and medial prefrontal cortex are responsible for the PPI-disruptive effect of dopamine D1 receptor agonists. These findings provide essential insights into the cellular and neural circuit mechanisms underlying the disruptive effects of dopamine D1 receptor agonists on PPI and may contribute to the development of novel treatments for neuropsychiatric diseases.


Assuntos
Benzazepinas , Agonistas de Dopamina , Camundongos Endogâmicos C57BL , Núcleo Accumbens , Córtex Pré-Frontal , Inibição Pré-Pulso , Receptores de Dopamina D1 , Animais , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Agonistas de Dopamina/farmacologia , Camundongos , Benzazepinas/farmacologia , Masculino , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Inibição Pré-Pulso/efeitos dos fármacos , Inibição Pré-Pulso/fisiologia , Reflexo de Sobressalto/efeitos dos fármacos , Filtro Sensorial/efeitos dos fármacos , Antagonistas de Dopamina/farmacologia
6.
J Affect Disord ; 356: 672-680, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38657771

RESUMO

BACKGROUND: Depression is a chronic psychiatric disorder related to diminished dopaminergic neurotransmission. Deep brain stimulation (DBS) has shown effectiveness in treating patients with treatment-refractory depression (TRD). This study aimed to evaluate the effect of DBS on dopamine D2 receptor binding in patients with TRD. METHODS: Six patients with TRD were treated with bed nucleus of the stria terminalis (BNST)-nucleus accumbens (NAc) DBS were recruited. Ultra-high sensitivity [11C]raclopride dynamic total-body positron emission tomography (PET) imaging was used to assess the brain D2 receptor binding. Each patient underwent a [11C]raclopride PET scan for 60-min under DBS OFF and DBS ON, respectively. A simplified reference tissue model was used to generate parametric images of binding potential (BPND) with the cerebellum as reference tissue. RESULTS: Depression and anxiety symptoms improved after 3-6 months of DBS treatment. Compared with two-day-nonstimulated conditions, one-day BNST-NAc DBS decreased [11C]raclopride BPND in the amygdala (15.9 %, p < 0.01), caudate nucleus (15.4 %, p < 0.0001) and substantia nigra (10.8 %, p < 0.01). LIMITATIONS: This study was limited to the small sample size and lack of a healthy control group. CONCLUSIONS: Chronic BNST-NAc DBS improved depression and anxiety symptoms, and short-term stimulation decreased D2 receptor binding in the amygdala, caudate nucleus, and substantia nigra. The findings suggest that DBS relieves depression and anxiety symptoms possibly by regulating the dopaminergic system.


Assuntos
Estimulação Encefálica Profunda , Transtorno Depressivo Resistente a Tratamento , Núcleo Accumbens , Tomografia por Emissão de Pósitrons , Racloprida , Receptores de Dopamina D2 , Humanos , Receptores de Dopamina D2/metabolismo , Estimulação Encefálica Profunda/métodos , Masculino , Feminino , Pessoa de Meia-Idade , Transtorno Depressivo Resistente a Tratamento/terapia , Transtorno Depressivo Resistente a Tratamento/metabolismo , Transtorno Depressivo Resistente a Tratamento/diagnóstico por imagem , Núcleo Accumbens/metabolismo , Núcleo Accumbens/diagnóstico por imagem , Adulto , Núcleos Septais/metabolismo , Núcleos Septais/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem , Resultado do Tratamento
7.
Proc Natl Acad Sci U S A ; 121(16): e2307982121, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38593084

RESUMO

A major aspiration of investors is to better forecast stock performance. Interestingly, emerging "neuroforecasting" research suggests that brain activity associated with anticipatory reward relates to market behavior and population-wide preferences, including stock price dynamics. In this study, we extend these findings to professional investors processing comprehensive real-world information on stock investment options while making predictions of long-term stock performance. Using functional MRI, we sampled investors' neural responses to investment cases and assessed whether these responses relate to future performance on the stock market. We found that our sample of investors could not successfully predict future market performance of the investment cases, confirming that stated preferences do not predict the market. Stock metrics of the investment cases were not predictive of future stock performance either. However, as investors processed case information, nucleus accumbens (NAcc) activity was higher for investment cases that ended up overperforming in the market. These findings remained robust, even when controlling for stock metrics and investors' predictions made in the scanner. Cross-validated prediction analysis indicated that NAcc activity could significantly predict future stock performance out-of-sample above chance. Our findings resonate with recent neuroforecasting studies and suggest that brain activity of professional investors may help in forecasting future stock performance.


Assuntos
Fenômenos Fisiológicos do Sistema Nervoso , Núcleo Accumbens , Humanos , Previsões , Investimentos em Saúde
8.
Behav Brain Res ; 466: 114983, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38580200

RESUMO

Humans and other animals exhibit aversive behavioral and emotional responses to unequal reward distributions compared with their conspecifics. Despite the significance of this phenomenon, experimental animal models designed to investigate social inequity aversion and delve into the underlying neurophysiological mechanisms are limited. In this study, we developed a rat model to determine the effects of socially equal or unequal reward and stress on emotional changes in male rats. During the training session, the rats were trained to escape when a sound cue was presented, and they were assigned to one of the following groups: all escaping rats [advantageous equity (AE)], freely moving rats alongside a restrained rat [advantageous inequity (AI)], all restrained rats [disadvantageous equity (DE)], and a rat restrained in the presence of freely moving companions [disadvantageous inequity (DI)]. During the test session, rats in the advantageous group (AE and AI) escaped after the cue sound (expected reward acquisition), whereas rats in the disadvantageous group (DE and DI) could not escape despite the cue being presented (expected reward deprivation). Emotional alteration induced by exposure to restraint stress under various social interaction circumstances was examined using an open field test. Notably, the DI group displayed reduced exploration of the center zone during the open field tests compared with the other groups, indicating heightened anxiety-like behaviors in response to reward inequity. Immunohistochemical analysis revealed increased c-Fos expression in the medial prefrontal and orbitofrontal cortices, coupled with reduced c-Fos expression in the striatum and nucleus accumbens under DI conditions, in contrast to the other experimental conditions. These findings provide compelling evidence that rats are particularly sensitive to reward inequity, shedding light on the neurophysiological basis for distinct cognitive processes that manifest when individuals are exposed to social equity and inequity situations.


Assuntos
Comportamento Animal , Emoções , Proteínas Proto-Oncogênicas c-fos , Recompensa , Estresse Psicológico , Animais , Masculino , Ratos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Emoções/fisiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Comportamento Animal/fisiologia , Comportamento Social , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/metabolismo , Sinais (Psicologia) , Ratos Sprague-Dawley
9.
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
10.
Biomolecules ; 14(4)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38672476

RESUMO

The recent approval of formulations of the endogenous neurosteroid allopregnanolone (brexanolone) and the synthetic neuroactive steroid SAGE-217 (zuranolone) to treat postpartum depression (PPD) has encouraged further research to elucidate why these potent enhancers of GABAAR function are clinically effective in this condition. Dopaminergic projections from the ventral tegmental area (VTA) to the nucleus accumbens are associated with reward/motivation and brain imaging studies report that individuals with PPD show reduced activity of this pathway in response to reward and infant engagement. However, the influence of neurosteroids on GABA-ergic transmission in the nucleus accumbens has received limited attention. Here, we investigate, in the medium spiny neurons (MSNs) of the mouse nucleus accumbens core, the effect of allopregnanolone, SAGE-217 and other endogenous and synthetic steroids of interest on fast phasic and tonic inhibition mediated by synaptic (α1/2ßγ2) and extrasynaptic (α4ßδ) GABAARs, respectively. We present evidence suggesting the resident tonic current results from the spontaneous opening of δ-GABAARs, where the steroid-enhanced tonic current is GABA-dependent. Furthermore, we demonstrate local neurosteroid synthesis in the accumbal slice preparation and reveal that GABA-ergic neurotransmission of MSNs is influenced by an endogenous neurosteroid tone. Given the dramatic fluctuations in allopregnanolone levels during pregnancy and postpartum, this neurosteroid-mediated local fine-tuning of GABAergic transmission in the MSNs will probably be perturbed.


Assuntos
Neuroesteroides , Núcleo Accumbens , Pregnanolona , Receptores de GABA-A , Animais , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Camundongos , Receptores de GABA-A/metabolismo , Neuroesteroides/metabolismo , Pregnanolona/farmacologia , Pregnanolona/metabolismo , Sinapses/metabolismo , Sinapses/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Feminino , Masculino , Transmissão Sináptica/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos
11.
Brain Res Bull ; 211: 110935, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38570076

RESUMO

Chronic ethanol consumption can lead to increased extracellular glutamate concentrations in key reward brain regions, such as medial prefrontal cortex (mPFC) and nucleus accumbens (NAc), and consequently leading to oxidative stress and neuroinflammation. Previous studies from our lab tested ß-lactam antibiotics and novel beta-lactam non-antibiotic, MC-100093, and showed these ß-lactam upregulated the major astrocytic glutamate transporter, GLT-1, and consequently reduced ethanol intake and normalized glutamate homeostasis. This present study tested the effects of novel synthetic ß-lactam non-antibiotic drug, MC-100093, in chronic ethanol intake and neuroinflammatory and trophic factors in subregions of the NAc (NAc core and shell) and mPFC (Prelimbic, PL; and Infralimbic, IL) of male P rats. MC-100093 treatment reduced ethanol intake after 5-week drinking regimen. Importantly, MC-100093 attenuated ethanol-induced downregulation of brain derived neurotrophic factor (BDNF) expression in these brain regions. In addition, MC-100093 attenuated ethanol-induced upregulation of pro-inflammatory cytokines such as TNF-a and HMGB1 in all these brain regions. Furthermore, MC-100093 treatment attenuated ethanol-induced increase in RAGE in these brain regions. MC-100093 prevented neuroinflammation caused by ethanol intake as well as increased neurotrophic factor in mesocorticolimbic brain regions. MC-100093 treatment reduced ethanol intake and this behavioral effect was associated with attenuation of reduced trophic factors and increased pro-inflammatory factors. MC-100093 is considered a small molecule that may have potential therapeutic effects for the treatment of the effects of chronic exposure to ethanol.


Assuntos
Etanol , Transportador 2 de Aminoácido Excitatório , Núcleo Accumbens , Córtex Pré-Frontal , Animais , Masculino , Transportador 2 de Aminoácido Excitatório/metabolismo , Etanol/farmacologia , Ratos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Biomarcadores/metabolismo , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/tratamento farmacológico , Alcoolismo/tratamento farmacológico , Alcoolismo/metabolismo , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos
12.
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
13.
Immunity ; 57(4): 837-839, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38599175

RESUMO

Activation of the peripheral immune system contributes to stress-related neuropsychiatric symptoms. Recently in Nature, Cathomas et al. demonstrate that stress-induced social avoidance is mediated by monocyte-derived MMP8 that remodels the extracellular space of the nucleus accumbens.


Assuntos
Depressão , Monócitos , Estresse Psicológico , Núcleo Accumbens
14.
Prog Neurobiol ; 236: 102615, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38641041

RESUMO

The gut-brain peptide ghrelin and its receptor are established as a regulator of hunger and reward-processing. However, the recently recognized ghrelin receptor inverse agonist, liver-expressed antimicrobial peptide 2 (LEAP2), is less characterized. The present study aimed to elucidate LEAP2s central effect on reward-related behaviors through feeding and its mechanism. LEAP2 was administrated centrally in mice and effectively reduced feeding and intake of palatable foods. Strikingly, LEAP2s effect on feeding was correlated to the preference of the palatable food. Further, LEAP2 reduced the rewarding memory of high preference foods, and attenuated the accumbal dopamine release associated with palatable food exposure and eating. Interestingly, LEAP2 was widely expressed in the brain, and particularly in reward-related brain areas such as the laterodorsal tegmental area (LDTg). This expression was markedly altered when allowed free access to palatable foods. Accordingly, infusion of LEAP2 into LDTg was sufficient to transiently reduce acute palatable food intake. Taken together, the present results show that central LEAP2 has a profound effect on dopaminergic reward signaling associated with food and affects several aspects of feeding. The present study highlights LEAP2s effect on reward, which may have applications for obesity and other reward-related psychiatric and neurological disorders.


Assuntos
Dopamina , Ingestão de Alimentos , Camundongos Endogâmicos C57BL , Núcleo Accumbens , Recompensa , Animais , Dopamina/metabolismo , Masculino , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Ingestão de Alimentos/fisiologia , Camundongos , Comportamento Alimentar/fisiologia , Proteínas Sanguíneas , Peptídeos Catiônicos Antimicrobianos
15.
Science ; 384(6693): eadk6742, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38669575

RESUMO

Drugs of abuse are thought to promote addiction in part by "hijacking" brain reward systems, but the underlying mechanisms remain undefined. Using whole-brain FOS mapping and in vivo single-neuron calcium imaging, we found that drugs of abuse augment dopaminoceptive ensemble activity in the nucleus accumbens (NAc) and disorganize overlapping ensemble responses to natural rewards in a cell type-specific manner. Combining FOS-Seq, CRISPR-perturbation, and single-nucleus RNA sequencing, we identified Rheb as a molecular substrate that regulates cell type-specific signal transduction in NAc while enabling drugs to suppress natural reward consumption. Mapping NAc-projecting regions activated by drugs of abuse revealed input-specific effects on natural reward consumption. These findings characterize the dynamic, molecular and circuit basis of a common reward pathway, wherein drugs of abuse interfere with the fulfillment of innate needs.


Assuntos
Homeostase , Núcleo Accumbens , Recompensa , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Animais , Camundongos , Neurônios/metabolismo , Drogas Ilícitas/efeitos adversos , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo/genética , Masculino , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Transdução de Sinais , Transtornos Relacionados ao Uso de Substâncias , Análise de Célula Única , Cocaína/farmacologia , Cálcio/metabolismo
16.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38688901

RESUMO

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Assuntos
Adenosina , Núcleo Accumbens , Receptor A2A de Adenosina , Sono de Ondas Lentas , Animais , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Masculino , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Camundongos , Adenosina/metabolismo , Adenosina/farmacologia , Regulação Alostérica , Sono de Ondas Lentas/fisiologia , Sono de Ondas Lentas/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Luz , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Humanos , Agonistas do Receptor A2 de Adenosina/farmacologia
17.
Sci Signal ; 17(832): eadl4738, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38626009

RESUMO

Cocaine use disorder (CUD) is a chronic neuropsychiatric condition that results from enduring cellular and molecular adaptations. Among substance use disorders, CUD is notable for its rising prevalence and the lack of approved pharmacotherapies. The nucleus accumbens (NAc), a region that is integral to the brain's reward circuitry, plays a crucial role in the initiation and continuation of maladaptive behaviors that are intrinsic to CUD. Leveraging advancements in neuroproteomics, we undertook a proteomic analysis that spanned membrane, cytosolic, nuclear, and chromatin compartments of the NAc in a mouse model. The results unveiled immediate and sustained proteomic modifications after cocaine exposure and during prolonged withdrawal. We identified congruent protein regulatory patterns during initial cocaine exposure and reexposure after withdrawal, which contrasted with distinct patterns during withdrawal. Pronounced proteomic shifts within the membrane compartment indicated adaptive and long-lasting molecular responses prompted by cocaine withdrawal. In addition, we identified potential protein translocation events between soluble-nuclear and chromatin-bound compartments, thus providing insight into intracellular protein dynamics after cocaine exposure. Together, our findings illuminate the intricate proteomic landscape that is altered in the NAc by cocaine use and provide a dataset for future research toward potential therapeutics.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Camundongos , Animais , Núcleo Accumbens/metabolismo , Proteômica , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/genética , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/psicologia , Cromatina/metabolismo
18.
ACS Chem Neurosci ; 15(9): 1738-1754, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38613458

RESUMO

Iboga alkaloids, also known as coronaridine congeners, have shown promise in the treatment of alcohol and opioid use disorders. The objective of this study was to evaluate the effects of catharanthine and 18-methoxycoronaridine (18-MC) on dopamine (DA) transmission and cholinergic interneurons in the mesolimbic DA system, nicotine-induced locomotor activity, and nicotine-taking behavior. Utilizing ex vivo fast-scan cyclic voltammetry (FSCV) in the nucleus accumbens core of male mice, we found that catharanthine or 18-MC differentially inhibited evoked DA release. Catharanthine inhibition of evoked DA release was significantly reduced by both α4 and α6 nicotinic acetylcholine receptors (nAChRs) antagonists. Additionally, catharanthine substantially increased DA release more than vehicle during high-frequency stimulation, although less potently than an α4 nAChR antagonist, which confirms previous work with nAChR antagonists. Interestingly, while catharanthine slowed DA reuptake measured via FSCV ex vivo, it also increased extracellular DA in striatal dialysate from anesthetized mice in vivo in a dose-dependent manner. Superfusion of catharanthine or 18-MC inhibited the firing rate of striatal cholinergic interneurons in a concentration dependent manner, which are known to potently modulate presynaptic DA release. Catharanthine or 18-MC suppressed acetylcholine currents in oocytes expressing recombinant rat α6/α3ß2ß3 or α6/α3ß4 nAChRs. In behavioral experiments using male Sprague-Dawley rats, systemic administration of catharanthine or 18-MC blocked nicotine enhancement of locomotor activity. Importantly, catharanthine attenuated nicotine self-administration in a dose-dependent manner while having no effect on food reinforcement. Lastly, administration of catharanthine and nicotine together greatly increased head twitch responses, indicating a potential synergistic hallucinogenic effect. These findings demonstrate that catharanthine and 18-MC have similar, but not identical effects on striatal DA dynamics, striatal cholinergic interneuron activity and nicotine psychomotor effects.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina , Dopamina , Ibogaína , Ibogaína/análogos & derivados , Nicotina , Receptores Nicotínicos , Animais , Dopamina/metabolismo , Masculino , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/efeitos dos fármacos , Nicotina/farmacologia , Ibogaína/farmacologia , Camundongos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Camundongos Endogâmicos C57BL , Antagonistas Nicotínicos/farmacologia , Oócitos/efeitos dos fármacos , Agonistas Nicotínicos/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Autoadministração , Xenopus laevis , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Relação Dose-Resposta a Droga , Atividade Motora/efeitos dos fármacos
19.
J Neuroendocrinol ; 36(5): e13389, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38599683

RESUMO

Hunger increases the motivation for calorie consumption, often at the expense of low-taste appeal. However, the neural mechanisms integrating calorie-sensing with increased motivation for calorie consumption remain unknown. Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus sense hunger, and the ingestion of caloric solutions promotes dopamine release in the absence of sweet taste perception. Therefore, we hypothesised that metabolic-sensing of hunger by AgRP neurons would be essential to promote dopamine release in the nucleus accumbens in response to caloric, but not non-caloric solutions. Moreover, we examined whether metabolic sensing in AgRP neurons affected taste preference for bitter solutions under conditions of energy need. Here we show that impaired metabolic sensing in AgRP neurons attenuated nucleus accumbens dopamine release in response to sucrose, but not saccharin, consumption. Furthermore, metabolic sensing in AgRP neurons was essential to distinguish nucleus accumbens dopamine response to sucrose consumption when compared with saccharin. Under conditions of hunger, metabolic sensing in AgRP neurons increased the preference for sucrose solutions laced with the bitter tastant, quinine, to ensure calorie consumption, whereas mice with impaired metabolic sensing in AgRP neurons maintained a strong aversion to sucrose/quinine solutions despite ongoing hunger. In conclusion, we demonstrate normal metabolic sensing in AgRP neurons drives the preference for calorie consumption, primarily when needed, by engaging dopamine release in the nucleus accumbens.


Assuntos
Proteína Relacionada com Agouti , Dopamina , Núcleo Accumbens , Sacarose , Núcleo Accumbens/metabolismo , Animais , Dopamina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Camundongos , Masculino , Preferências Alimentares/fisiologia , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fome/fisiologia , Percepção Gustatória/fisiologia
20.
Nat Commun ; 15(1): 2543, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514654

RESUMO

Accumulating evidence points to dysregulations of the Nucleus Accumbens (NAc) in eating disorders (ED), however its precise contribution to ED symptomatic dimensions remains unclear. Using chemogenetic manipulations in male mice, we found that activity of dopamine D1 receptor-expressing neurons of the NAc core subregion facilitated effort for a food reward as well as voluntary exercise, but decreased food intake, while D2-expressing neurons have opposite effects. These effects are congruent with D2-neurons being more active than D1-neurons during feeding while it is the opposite during running. Chronic manipulations of each subpopulations had limited effects on energy balance. However, repeated activation of D1-neurons combined with inhibition of D2-neurons biased behavior toward activity-related energy expenditure, whilst the opposite manipulations favored energy intake. Strikingly, concomitant activation of D1-neurons and inhibition of D2-neurons precipitated weight loss in anorexia models. These results suggest that dysregulations of NAc dopaminoceptive neurons might be at the core of EDs.


Assuntos
Núcleo Accumbens , Receptores de Dopamina D2 , Camundongos , Masculino , Animais , Núcleo Accumbens/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Neurônios/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Metabolismo Energético
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA