Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.957
Filtrar
1.
Nature ; 630(8015): 141-148, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38778097

RESUMO

Fentanyl is a powerful painkiller that elicits euphoria and positive reinforcement1. Fentanyl also leads to dependence, defined by the aversive withdrawal syndrome, which fuels negative reinforcement2,3 (that is, individuals retake the drug to avoid withdrawal). Positive and negative reinforcement maintain opioid consumption, which leads to addiction in one-fourth of users, the largest fraction for all addictive drugs4. Among the opioid receptors, µ-opioid receptors have a key role5, yet the induction loci of circuit adaptations that eventually lead to addiction remain unknown. Here we injected mice with fentanyl to acutely inhibit γ-aminobutyric acid-expressing neurons in the ventral tegmental area (VTA), causing disinhibition of dopamine neurons, which eventually increased dopamine in the nucleus accumbens. Knockdown of µ-opioid receptors in VTA abolished dopamine transients and positive reinforcement, but withdrawal remained unchanged. We identified neurons expressing µ-opioid receptors in the central amygdala (CeA) whose activity was enhanced during withdrawal. Knockdown of µ-opioid receptors in CeA eliminated aversive symptoms, suggesting that they mediate negative reinforcement. Thus, optogenetic stimulation caused place aversion, and mice readily learned to press a lever to pause optogenetic stimulation of CeA neurons that express µ-opioid receptors. Our study parses the neuronal populations that trigger positive and negative reinforcement in VTA and CeA, respectively. We lay out the circuit organization to develop interventions for reducing fentanyl addiction and facilitating rehabilitation.


Assuntos
Neurônios Dopaminérgicos , Fentanila , Núcleo Accumbens , Receptores Opioides mu , Reforço Psicológico , Síndrome de Abstinência a Substâncias , Área Tegmentar Ventral , Animais , Fentanila/farmacologia , Receptores Opioides mu/metabolismo , Camundongos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/fisiologia , Masculino , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Dopamina/metabolismo , Optogenética , Núcleo Central da Amígdala/metabolismo , Núcleo Central da Amígdala/efeitos dos fármacos , Núcleo Central da Amígdala/fisiologia , Feminino , Camundongos Endogâmicos C57BL , Transtornos Relacionados ao Uso de Opioides/metabolismo , Analgésicos Opioides/farmacologia , Analgésicos Opioides/administração & dosagem
2.
Addict Biol ; 29(5): e13403, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38735880

RESUMO

Synthetic opioids such as fentanyl contribute to the vast majority of opioid-related overdose deaths, but fentanyl use remains broadly understudied. Like other substances with misuse potential, opioids cause lasting molecular adaptations to brain reward circuits, including neurons in the ventral tegmental area (VTA). The VTA contains numerous cell types that play diverse roles in opioid use and relapse; however, it is unknown how fentanyl experience alters the transcriptional landscape in specific subtypes. Here, we performed single nuclei RNA sequencing to study transcriptional programs in fentanyl-experienced mice. Male and female C57/BL6 mice self-administered intravenous fentanyl (1.5 µg/kg/infusion) or saline for 10 days. After 24 h abstinence, VTA nuclei were isolated and prepared for sequencing on the 10× platform. We identified different patterns of gene expression across cell types. In dopamine neurons, we found enrichment of genes involved in growth hormone signalling. In dopamine-glutamate-GABA combinatorial neurons, and some GABA neurons, we found enrichment of genes involved in Pi3k-Akt signalling. In glutamate neurons, we found enrichment of genes involved in cholinergic signalling. We identified transcriptional regulators for the differentially expressed genes in each neuron cluster, including downregulated transcriptional repressor Bcl6, and upregulated transcription factor Tcf4. We also compared the fentanyl-induced gene expression changes identified in mouse VTA with a published rat dataset in bulk VTA, and found overlap in genes related to GABAergic signalling and extracellular matrix interaction. Together, we provide a comprehensive picture of how fentanyl self-administration alters the transcriptional landscape of the mouse VTA that serves as the foundation for future mechanistic studies.


Assuntos
Analgésicos Opioides , Fentanila , Camundongos Endogâmicos C57BL , Área Tegmentar Ventral , Animais , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Camundongos , Fentanila/farmacologia , Masculino , Feminino , Analgésicos Opioides/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Autoadministração , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transtornos Relacionados ao Uso de Opioides/genética
3.
eNeuro ; 11(6)2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38806231

RESUMO

Amylin, a pancreatic hormone that is cosecreted with insulin, has been highlighted as a potential treatment target for obesity. Amylin receptors are distributed widely throughout the brain and are coexpressed on mesolimbic dopamine neurons. Activation of amylin receptors is known to reduce food intake, but the neurochemical mechanisms behind this remain to be elucidated. Amylin receptor activation in the ventral tegmental area (VTA), a key dopaminergic nucleus in the mesolimbic reward system, has a potent ability to suppress intake of palatable fat and sugar solutions. Although previous work has demonstrated that VTA amylin receptor activation can dampen mesolimbic dopamine signaling elicited by random delivery of sucrose, whether this is also the case for fat remains unknown. Herein we tested the hypothesis that amylin receptor activation in the VTA of male rats would attenuate dopamine signaling in the nucleus accumbens core in response to random intraoral delivery of either fat or sugar solutions. Results show that fat solution produces a greater potentiation of accumbens dopamine than an isocaloric sucrose solution. Moreover, activation of VTA amylin receptors elicits a more robust suppression of accumbens dopamine signaling in response to fat solution than to sucrose. Taken together these results shed new light on the amylin system as a therapeutic target for obesity and emphasize the reinforcing nature of high-fat/high-sugar diets.


Assuntos
Dopamina , Núcleo Accumbens , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas , Área Tegmentar Ventral , Animais , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Masculino , Dopamina/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas/metabolismo , Ratos Sprague-Dawley , Gorduras na Dieta/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Agonistas dos Receptores da Amilina/farmacologia , Ratos , Sacarose/administração & dosagem , Sacarose/farmacologia
4.
Int J Mol Sci ; 25(10)2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38791298

RESUMO

Tobacco use disorder represents a significant public health challenge due to its association with various diseases. Despite awareness efforts, smoking rates remain high, partly due to ineffective cessation methods and the spread of new electronic devices. This study investigated the impact of prolonged nicotine exposure via a heat-not-burn (HnB) device on selected genes and signaling proteins involved in inflammatory processes in the rat ventral tegmental area (VTA) and nucleus accumbens (NAc), two brain regions associated with addiction to different drugs, including nicotine. The results showed a reduction in mRNA levels for PPARα and PPARγ, two nuclear receptors and anti-inflammatory transcription factors, along with the dysregulation of gene expression of the epigenetic modulator KDM6s, in both investigated brain areas. Moreover, decreased PTEN mRNA levels and higher AKT phosphorylation were detected in the VTA of HnB-exposed rats with respect to their control counterparts. Finally, significant alterations in ERK 1/2 phosphorylation were observed in both mesolimbic areas, with VTA decrease and NAc increase, respectively. Overall, the results suggest that HnB aerosol exposure disrupts intracellular pathways potentially involved in the development and maintenance of the neuroinflammatory state. Moreover, these data highlight that, similar to conventional cigarettes, HnB devices use affects specific signaling pathways shaping neuroinflammatory process in the VTA and NAc, thus triggering mechanisms that are currently considered as potentially relevant for the development of addictive behavior.


Assuntos
Núcleo Accumbens , Área Tegmentar Ventral , Animais , Ratos , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Masculino , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/etiologia , PPAR gama/metabolismo , PPAR gama/genética , Transdução de Sinais/efeitos dos fármacos , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Fumaça/efeitos adversos , Nicotina/efeitos adversos , Ratos Wistar , Nicotiana/efeitos adversos , Tabagismo/metabolismo , Fosforilação/efeitos dos fármacos
5.
Behav Brain Res ; 468: 115040, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38723675

RESUMO

Neurotoxins have been extensively investigated, particularly in the field of neuroscience. They induce toxic damage, oxidative stress, and inflammation on neurons, triggering neuronal dysfunction and neurodegenerative diseases. Here we demonstrate the neuroprotective effect of a silicon (Si)-based hydrogen-producing agent (Si-based agent) in a juvenile neurotoxic mouse model induced by 6-hydroxydopamine (6-OHDA). The Si-based agent produces hydrogen in bowels and functions as an antioxidant and anti-inflammatory agent. However, the effects of the Si-based agent on neural degeneration in areas other than the lesion and behavioral alterations caused by it are largely unknown. Moreover, the neuroprotective effects of Si-based agent in the context of lactation and use during infancy have not been explored in prior studies. In this study, we show the neuroprotective effect of the Si-based agent on 6-OHDA during lactation period and infancy using the mouse model. The Si-based agent safeguards against the degradation and neuronal cell death of dopaminergic neurons and loss of dopaminergic fibers in the striatum (STR) and ventral tegmental area (VTA) caused by 6-OHDA. Furthermore, the Si-based agent exhibits a neuroprotective effect on the length of axon initial segment (AIS) in the layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC). As a result, the Si-based agent mitigates hyperactive behavior in a juvenile neurotoxic mouse model induced by 6-OHDA. These results suggest that the Si-based agent serves as an effective neuroprotectant and antioxidant against neurotoxic effects in the brain, offering the possibility of the Si-based agent as a neuroprotectant for nervous system diseases.


Assuntos
Modelos Animais de Doenças , Neurônios Dopaminérgicos , Hidrogênio , Fármacos Neuroprotetores , Oxidopamina , Silício , Animais , Fármacos Neuroprotetores/farmacologia , Oxidopamina/farmacologia , Camundongos , Silício/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Hidrogênio/farmacologia , Hidrogênio/administração & dosagem , Masculino , Síndromes Neurotóxicas/tratamento farmacológico , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Camundongos Endogâmicos C57BL
6.
Alcohol Alcohol ; 59(4)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38742547

RESUMO

AIMS: Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD: We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT: Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS: Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.


Assuntos
Consumo de Bebidas Alcoólicas , Neurônios , Quinina , Caracteres Sexuais , Animais , Quinina/farmacologia , Feminino , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/efeitos dos fármacos , Córtex Insular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Etanol/farmacologia , Ácido Glutâmico/metabolismo
7.
Bipolar Disord ; 26(4): 376-387, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38558302

RESUMO

BACKGROUND: Treatment of refractory bipolar disorder (BD) is extremely challenging. Deep brain stimulation (DBS) holds promise as an effective treatment intervention. However, we still understand very little about the mechanisms of DBS and its application on BD. AIM: The present study aimed to investigate the behavioural and neurochemical effects of ventral tegmental area (VTA) DBS in an animal model of mania induced by methamphetamine (m-amph). METHODS: Wistar rats were given 14 days of m-amph injections, and on the last day, animals were submitted to 20 min of VTA DBS in two different patterns: intermittent low-frequency stimulation (LFS) or continuous high-frequency stimulation (HFS). Immediately after DBS, manic-like behaviour and nucleus accumbens (NAc) phasic dopamine (DA) release were evaluated in different groups of animals through open-field tests and fast-scan cyclic voltammetry. Levels of NAc dopaminergic markers were evaluated by immunohistochemistry. RESULTS: M-amph induced hyperlocomotion in the animals and both DBS parameters reversed this alteration. M-amph increased DA reuptake time post-sham compared to baseline levels, and both LFS and HFS were able to block this alteration. LFS was also able to reduce phasic DA release when compared to baseline. LFS was able to increase dopamine transporter (DAT) expression in the NAc. CONCLUSION: These results demonstrate that both VTA LFS and HFS DBS exert anti-manic effects and modulation of DA dynamics in the NAc. More specifically the increase in DA reuptake driven by increased DAT expression may serve as a potential mechanism by which VTA DBS exerts its anti-manic effects.


Assuntos
Estimulação Encefálica Profunda , Modelos Animais de Doenças , Mania , Metanfetamina , Ratos Wistar , Área Tegmentar Ventral , Animais , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Metanfetamina/farmacologia , Masculino , Ratos , Mania/terapia , Mania/induzido quimicamente , Estimulantes do Sistema Nervoso Central/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Transtorno Bipolar/terapia , Transtorno Bipolar/induzido quimicamente
8.
Eur J Neurosci ; 59(12): 3422-3444, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38679044

RESUMO

Drug dependence is characterized by a switch in motivation wherein a positively reinforcing substance can become negatively reinforcing. Put differently, drug use can transform from a form of pleasure-seeking to a form of relief-seeking. Ventral tegmental area (VTA) GABA neurons form an anatomical point of divergence between two double dissociable pathways that have been shown to be functionally implicated and necessary for these respective motivations to seek drugs. The tegmental pedunculopontine nucleus (TPP) is necessary for opiate conditioned place preferences (CPP) in previously drug-naïve rats and mice, whereas dopaminergic (DA) transmission in the nucleus accumbens (NAc) is necessary for opiate CPP in opiate-dependent and withdrawn (ODW) rats and mice. Here, we show that this switch in functional anatomy is contingent upon the gap junction-forming protein, connexin-36 (Cx36), in VTA GABA neurons. Intra-VTA infusions of the Cx36 blocker, mefloquine, in ODW rats resulted in a reversion to a drug-naïve-like state wherein the TPP was necessary for opiate CPP and where opiate withdrawal aversions were lost. Consistent with these data, conditional knockout mice lacking Cx36 in GABA neurons (GAD65-Cre;Cx36 fl(CFP)/fl(CFP)) exhibited a perpetual drug-naïve-like state wherein opiate CPP was always DA independent, and opiate withdrawal aversions were absent even in mice subjected to an opiate dependence and withdrawal induction protocol. Further, viral-mediated rescue of Cx36 in VTA GABA neurons was sufficient to restore their susceptibility to an ODW state wherein opiate CPP was DA dependent. Our findings reveal a functional role for VTA gap junctions that has eluded prevailing circuit models of addiction.


Assuntos
Conexinas , Neurônios GABAérgicos , Proteína delta-2 de Junções Comunicantes , Junções Comunicantes , Transtornos Relacionados ao Uso de Opioides , Área Tegmentar Ventral , Animais , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Conexinas/metabolismo , Conexinas/genética , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/efeitos dos fármacos , Junções Comunicantes/metabolismo , Junções Comunicantes/efeitos dos fármacos , Masculino , Ratos , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Mefloquina/farmacologia , Camundongos , Ratos Sprague-Dawley , Núcleo Tegmental Pedunculopontino/metabolismo , Núcleo Tegmental Pedunculopontino/efeitos dos fármacos
9.
Exp Neurol ; 376: 114758, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38513970

RESUMO

Impaired long-term memory, a complication of traumatic stress including hemorrhage shock and resuscitation (HSR), has been reported to be associated with multiple neurodegenerations. The ventral tegmental area (VTA) participates in both learned appetitive and aversive behaviors. In addition to being prospective targets for the therapy of addiction, depression, and other stress-related diseases, VTA glutamatergic neurons are becoming more widely acknowledged as powerful regulators of reward and aversion. This study revealed that HSR exposure induces memory impairments and decreases the activation in glutamatergic neurons, and decreased ß power in the VTA. We also found that optogenetic activation of glutamatergic neurons in the VTA mitigated HSR-induced memory impairments, and restored ß power. Moreover, hydrogen sulfide (H2S), a gasotransmitter with pleiotropic roles, has neuroprotective functions at physiological concentrations. In vivo, H2S administration improved HSR-induced memory deficits, elevated c-fos-positive vesicular glutamate transporters (Vglut2) neurons, increased ß power, and restored the balance of γ-aminobutyric acid (GABA) and glutamate in the VTA. This work suggests that glutamatergic neuron stimulation via optogenetic assay and exogenous H2S may be useful therapeutic approaches for improving memory deficits following HSR.


Assuntos
Modelos Animais de Doenças , Ácido Glutâmico , Sulfeto de Hidrogênio , Transtornos da Memória , Camundongos Endogâmicos C57BL , Neurônios , Animais , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/uso terapêutico , Camundongos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Transtornos da Memória/terapia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Choque Hemorrágico , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Optogenética/métodos
10.
Life Sci ; 328: 121900, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37391066

RESUMO

AIMS: Epidemiological evidence suggests that comorbidity of obesity and depression is extremely common and continues to grow in prevalence. However, the mechanisms connecting these two conditions are unknown. In this study, we explored how treatment with KATP channel blocker glibenclamide (GB) or the well-known metabolic regulator FGF21 impact male mice with high-fat diet (HFD)-induced obesity and depressive-like behaviors. MATERIALS AND METHODS: Mice were fed with HFD for 12 weeks and then treated with recombinant FGF21 protein by infusion for 2 weeks, followed by intraperitoneal injection of 3 mg/kg recombinant FGF21 once per day for 4 days. Measurements were made of catecholamine levels, energy expenditure, biochemical endpoints and behavior tests, including sucrose preference and forced swim tests were. Alternatively, animals were infused with GB into brown adipose tissue (BAT). The WT-1 brown adipocyte cell line was used for molecular studies. KEY FINDINGS: Compared to HFD controls, HFD + FGF21 mice exhibited less severe metabolic disorder symptoms, improved depressive-like behaviors, and more extensive mesolimbic dopamine projections. FGF21 treatment also rescued HFD-induced dysregulation of FGF21 receptors (FGFR1 and co-receptor ß-klotho) in the ventral tegmental area (VTA), and it altered dopaminergic neuron activity and morphology in HFD-fed mice. Importantly, we also found that FGF21 mRNA level and FGF21 release were increased in BAT after administration of GB, and GB treatment to BAT reversed HFD-induced dysregulation of FGF21 receptors in the VTA. SIGNIFICANCE: GB administration to BAT stimulates FGF21 production in BAT, corrects HFD-induced dysregulation of FGF21 receptor dimers in VTA dopaminergic neurons, and attenuates depression-like symptoms.


Assuntos
Tecido Adiposo Marrom , Depressão , Fatores de Crescimento de Fibroblastos , Glibureto , Hipoglicemiantes , Obesidade , Animais , Masculino , Camundongos , Tecido Adiposo Marrom/efeitos dos fármacos , Depressão/complicações , Depressão/tratamento farmacológico , Dieta Hiperlipídica , Fatores de Crescimento de Fibroblastos/administração & dosagem , Fatores de Crescimento de Fibroblastos/genética , Glibureto/administração & dosagem , Hipoglicemiantes/administração & dosagem , Doenças Metabólicas/tratamento farmacológico , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/patologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/patologia , Proteínas Recombinantes/administração & dosagem
11.
Behav Brain Res ; 440: 114249, 2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36496077

RESUMO

In the present study, we investigated the effects of the glucagon-like peptide-1 (GLP-1) agonist exendin-4 (Ex-4) on the stimulatory action of neuropeptide Y (NPY) and ghrelin. These effects were examined in relation to operant responding for palatable food or voluntary ethanol intake in a two-bottle limited access paradigm. Male Sprague Dawley rats, each with ventral tegmental area (VTA) unilateral guide cannulae, were used. Ex-4 was paired with either NPY, ghrelin, or combined NPY and ghrelin treatment. Our results indicated that while NPY and ghrelin reliably stimulated operant responding for sucrose pellets and increased ethanol intake, Ex-4 suppressed intake and, most importantly, significantly reduced the effects of NPY and ghrelin. Overall, this work provides compelling evidence that VTA GLP-1, NPY, and ghrelin systems interact within the brain to modulate reward salience.


Assuntos
Exenatida , Grelina , Neuropeptídeo Y , Recompensa , Área Tegmentar Ventral , Animais , Masculino , Ratos , Etanol/farmacologia , Exenatida/farmacologia , Grelina/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Neuropeptídeo Y/farmacologia , Ratos Sprague-Dawley , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
12.
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
13.
Neuropharmacology ; 206: 108940, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-34982973

RESUMO

The interaction among the acetylcholine (ACh)-ergic REM-ON neurons in the pedunculo-pontine area (PPT), noradrenergic REM-OFF neurons in locus coeruleus (LC) and GABA-ergic neurons in the regulation of rapid eye movement sleep (REMS) have been studied in relative details; however, many questions including the role of dopamine (DA) remain unanswered. The ventral tegmental area (VTA) is rich in DA-ergic neurons, which have been implicated with schizophrenia and depression, when REMS is significantly affected. Also, some of the symptoms of REMS and these diseases are common. As the ACh-ergic REM-ON neurons in the PPT project to VTA, we proposed that such inputs might affect REMS, dreams and hallucinations. We recorded sleep-wake-REMS in freely moving, chronically prepared rats under three controlled experimental conditions. In different sets of experiments, either the ACh-ergic inputs to the VTA were blocked by local microinjection of Scopolamine (Scop) alone, or, the PPT neurons were bilaterally stimulated by Glutamate (Glut), or, the PPT neurons were stimulated by Glut in presence of Scop into the VTA. It was observed that Glut into PPT and Scop into the VTA significantly increased and decreased REMS, respectively. Additionally, PPT stimulation induced increased REMS was prevented in the presence of Scop into the VTA. Based on these findings we propose that inputs from ACh-ergic REM-ON neurons to VTA increase REMS and it could be a possible circuitry for expressions of hallucinations and dreams.


Assuntos
Neurônios Colinérgicos/fisiologia , Neurônios Dopaminérgicos/fisiologia , Tegmento Pontino/fisiologia , Sono REM/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Antagonistas Colinérgicos/farmacologia , Ratos , Escopolamina/farmacologia , Sono REM/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
14.
Mol Neurobiol ; 59(1): 35-46, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34618330

RESUMO

We recently reported that intraperitoneal injection of 7,8-dihydroxyflavone (7,8-DHF), a brain-derived neurotrophic factor-mimicking small compound, could attenuate alcohol-related behaviors in a two-bottle choice ethanol consumption procedure (IA2BC) in rats via tropomyosin receptor kinase B in the ventral tegmental area (VTA), which is closely related to alcohol use disorder. However, the detailed mechanisms underlying the regulation of 7,8-DHF on alcohol drinking behavior remain elusive. In this study, we determined the role of nitric oxide (NO), a pleiotropic signaling molecule, in the VTA in the action of 7,8-DHF upon alcohol drinking behavior. Intermittent alcohol exposure led to the overexpression of NO in the VTA, especially 72 h after withdrawal from four weeks of ethanol exposure in IA2BC rats. A higher amount of alcohol intake was also found at the same time point, consistent with the overexpression of NO in the VTA. Microinjection of NG-Nitro-l-Arginine Methyl Ester, (NO synthase inhibitor) or 2-4-carboxyphenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (NO scavenger) into the VTA inhibited alcohol intake, whereas application of S-Nitroso-N-acetyl-DL-penicillamine (SNAP, the NO donor) in the VTA further enhanced alcohol consumption in IA2BC rats. Interestingly, either 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one (a sGC inhibitor) or KT5823 [a selective protein kinase G (PKG) inhibitor] blocked NO's enhancing effect on ethanol intake. Intraperitoneal injection of 7,8-DHF reduced the overexpression of NO; SNAP microinjected into the VTA reversed the inhibitory effects of 7,8-DHF on alcohol consumption. Our findings suggest that NO-cGMP-PKG might be involved in regulation of 7,8-DHF on alcohol consumption in IA2BC rats.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Flavonas/farmacologia , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , GMP Cíclico/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Área Tegmentar Ventral/metabolismo
15.
Behav Brain Res ; 417: 113596, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-34562552

RESUMO

Cotinine is the major metabolite of nicotine and has recently been shown to be self-administered intravenously by rats. However, mechanisms underlying cotinine self-administration remained unknown. Mesolimbic dopamine system projecting from the ventral tegmental area (VTA) to nucleus accumbens (NAC) is closely implicated in drug reinforcement, including nicotine. The objective of the current study was to determine potential involvement of mesolimbic dopamine system in cotinine self-administration. An intracranial self-administration experiment demonstrates that cotinine at 0.88 and 1.76 ng/100 nl/infusion was self-infused into the VTA by rats. Rats produced more infusions of cotinine than vehicle and responded more on active than inactive lever during acquisition, reduced responding when cotinine was replaced by vehicle, and resumed responding during re-exposure to cotinine. Microinjection of cotinine at 1.76 ng/100 nl/infusion into the VTA increased extracellular dopamine levels within the NAC. Subcutaneous injection of cotinine at 1 mg/kg also increased extracellular dopamine levels within the NAC. Administration of the D1-like receptor antagonist SCH 23390 attenuated intravenous cotinine self-administration. On the other hand, bupropion, a catecholamine uptake inhibitor, did not significantly alter intravenous cotinine self-administration. These results suggest that activation of mesolimbic dopamine system may represent one cellular mechanism underlying cotinine self-administration. This shared mechanism between cotinine and nicotine suggests that cotinine may play a role in nicotine reinforcement.


Assuntos
Cotinina/administração & dosagem , Dopamina/fisiologia , Sistema Límbico/fisiologia , Autoadministração , Animais , Benzazepinas/antagonistas & inibidores , Encéfalo/metabolismo , Bupropiona , Inibidores da Captação de Dopamina , Sistema Límbico/efeitos dos fármacos , Masculino , Microinjeções , Núcleo Accumbens/efeitos dos fármacos , Ratos , Reforço Psicológico , Área Tegmentar Ventral/efeitos dos fármacos
16.
Artigo em Inglês | MEDLINE | ID: mdl-34339759

RESUMO

Methylphenidate (MPH) is a drug routinely used for patients with attention deficit and hyperactivity disorder (ADHD). Concerns arise about psychostimulant use, with dramatic increases in prescriptions. Besides, antipsychotic drugs are often administered in combination with MPH. In this study, we examine the consequences of MPH exposure in combination with dopamine D2 receptor antagonism (eticlopride) on midbrain dopaminergic neurons in anaesthetised rodents, using in vivo extracellular single-cell electrophysiology. As expected, we show that methylphenidate (2 mg/kg, i.v.) decreases the firing and bursting activities of ventral tegmental area (VTA) dopamine neurons, an effect that is reversed with eticlopride (0.2 mg/kg, i.v.). However, using such a paradigm, we observed higher firing and bursting activities than under baseline conditions. Furthermore, we demonstrate that such an effect is dependent on dual alpha-1 and dopamine D1 receptors, as well as glutamatergic transmission, through glutamate N-Methyl-D-aspartate (NMDA) receptor activation. Chronic MPH treatment during adolescence greatly dampens MPH-induced excitatory effects measured at adulthood. To conclude, we demonstrated here that a combination of methylphenidate and a dopamine D2 receptor antagonist produced long-lasting consequences on midbrain dopamine neurons, via glutamatergic-dependent mechanisms.


Assuntos
Inibidores da Captação de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Eletrofisiologia , Metilfenidato/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Modelos Animais de Doenças , Antagonistas de Dopamina/administração & dosagem , Neurônios Dopaminérgicos/citologia , Quimioterapia Combinada , Masculino , Mesencéfalo , Ratos , Receptores Dopaminérgicos , Receptores de N-Metil-D-Aspartato/fisiologia , Salicilamidas/administração & dosagem
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.
Neuropharmacology ; 202: 108859, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710468

RESUMO

Nicotine, the addictive component of tobacco, has bivalent rewarding and aversive properties. Recently, the lateral habenula (LHb), a structure that controls ventral tegmental area (VTA) dopamine (DA) function, has attracted attention as it is potentially involved in the aversive properties of drugs of abuse. Hitherto, the LHb-modulation of nicotine-induced VTA neuronal activity in vivo is unknown. Using standard single-extracellular recording in anesthetized rats, we observed that intravenous administration of nicotine hydrogen tartrate (25-800 µg/kg i.v.) caused a dose-dependent increase in the basal firing rate of the LHb neurons of nicotine-naïve rats. This effect underwent complete desensitization in chronic nicotine (6 mg/kg/day for 14 days)-treated animals. As previously reported, acute nicotine induced an increase in the VTA DA neuronal firing rate. Interestingly, only neurons located medially (mVTA) but not laterally (latVTA) within the VTA were responsive to acute nicotine. This pattern of activation was reversed by chronic nicotine exposure which produced the selective increase of latVTA neuronal activity. Acute lesion of the LHb, similarly to chronic nicotine treatment, reversed the pattern of DA cell activation induced by acute nicotine increasing latVTA but not mVTA neuronal activity. Our evidence indicates that LHb plays an important role in mediating the effects of acute and chronic nicotine within the VTA by activating distinct subregional responses of DA neurons. The LHb/VTA modulation might be part of the neural substrate of nicotine aversive properties. By silencing the LHb chronic nicotine could shift the balance of motivational states toward the reward.


Assuntos
Dopamina/fisiologia , Eletroencefalografia/métodos , Habenula/efeitos dos fármacos , Habenula/fisiopatologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiopatologia , Nicotina/efeitos adversos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiopatologia , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Relação Dose-Resposta a Droga , Masculino , Nicotina/farmacologia , Ratos Sprague-Dawley , Recompensa
19.
Artigo em Inglês | MEDLINE | ID: mdl-34454991

RESUMO

The hyperpolarization-activated cation current (Ih) is a determinant of intrinsic excitability in various cells, including dopaminergic neurons (DA) of the ventral tegmental area (VTA). In contrast to other cellular conductances, Ih is activated by hyperpolarization negative to -55 mV and activating Ih produces a time-dependent depolarizing current. Our laboratory demonstrated that cocaine sensitization, a chronic cocaine behavioral model, significantly reduces Ih amplitude in VTA DA neurons. Despite this reduction in Ih, the spontaneous firing of VTA DA cells after cocaine sensitization remained similar to control groups. Although the role of Ih in controlling VTA DA excitability is still poorly understood, our hypothesis is that Ih reduction could play a role of a homeostatic controller compensating for cocaine-induced change in excitability. Using in vivo single-unit extracellular electrophysiology in isoflurane anesthetized rats, we explored the contribution of Ih on spontaneous firing patterns of VTA DA neurons. A key feature of spontaneous excitability is bursting activity; bursting is defined as trains of two or more spikes occurring within a short interval and followed by a prolonged period of inactivity. Burst activity increases the reliability of information transfer. To elucidate the contribution of Ih to spontaneous firing patterns of VTA DA neurons, we locally infused an Ih blocker (ZD 7288, 8.3 µM) and evaluated its effect. Ih blockade significantly reduced firing rate, bursting frequency, and percent of spikes within a burst. In addition, Ih blockade significantly reduced acute cocaine-induced spontaneous firing rate, bursting frequency, and percent of spikes within a burst. Using whole-cell patch-clamp, we determine the progressive reduction of Ih after acute and chronic cocaine administration (15 mg/k.g intraperitoneally). Our data show a significant reduction (~25%) in Ih amplitude after 24 but not 2 h of acute cocaine administration. These results suggest that a progressive reduction of Ih could serve as a homeostatic regulator of cocaine-induced spontaneous firing patterns related to VTA DA excitability.


Assuntos
Cocaína/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Área Tegmentar Ventral/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Eletrofisiologia , Masculino , Ratos
20.
Addict Biol ; 27(2): e13120, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34825430

RESUMO

Dopamine neurons in the substantia nigra (SN) and ventral tegmental area (VTA) play a central role in the reinforcing properties of abused drugs including methamphetamine and cocaine. Chronic effects of psychostimulants in the SN/VTA also involve non-dopaminergic transmitters, including glutamate and the stress-related peptide corticotropin-releasing factor (CRF). In the SN/VTA, astrocytes express a variety of membrane-bound neurotransmitter receptors and transporters that influence neurotransmission. CRF receptor type 2 (CRF2) activity in the VTA is important for stress-induced relapse and drug-seeking behaviour, but the localization of its effects is incompletely understood. Here, we first identified CRF2 transcript in astrocytes of the SN/VTA using RNA-Seq in Aldh1l1;NuTRAP mice and confirmed it using in situ hybridization (RNAscope) in wild-type mice. We then used immunofluorescence to quantify the astrocytic marker protein S100ß, glial-specific glutamate/aspartate transporter GLAST, and CRF2 in the SN/VTA following 12 days of treatment (i.p.) with methamphetamine (3 mg/kg), cocaine (10 mg/kg), or saline. We observed a significant decrease in GLAST immunofluorescence in brains of psychostimulant treated mice compared with saline controls. In addition, we observed increased labelling of CRF2 in drug treated groups, a decrease in the number of S100ß positive cells, and an increase of co-staining of CRF2 with both S100ß and tyrosine hydroxylase (dopamine neurons). Our results suggest a significant interaction between CRF2, GLAST, and astrocytes in the midbrain that emerges with repeated exposure to psychostimulants. These findings provide rationale for future investigation of astrocyte-based strategies for altering cellular and circuit function in response to stress and drug exposure.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Cocaína , Hormônio Liberador da Corticotropina/metabolismo , Metanfetamina , Área Tegmentar Ventral , Animais , Astrócitos/metabolismo , Cocaína/farmacologia , Metanfetamina/farmacologia , Camundongos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...