Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 432
Filtrar
1.
Nutrients ; 16(18)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39339647

RESUMEN

Nicotine dependence is an important cause of excessive exposure to tobacco combustion compounds in most smokers. Nicotine replacement therapy is the main method to treat nicotine dependence, but it still has its shortcomings, such as the inability to mitigate withdrawal effects and limited applicability. It has been hypothesized that a combination of low-dose nicotine and caffeine could achieve the same psychological stimulation effect as a high dose of nicotine without causing nicotine withdrawal effects. To establish a model of nicotine dependence, male C57BL/6J mice were subcutaneously injected four times a day with nicotine (2 mg/kg) for 15 days and fed with water containing nicotine at the same time. They were randomly divided into four groups. After 24 h of withdrawal, different groups were injected with saline, nicotine (0.25 mg/kg or 0.1 mg/kg), or nicotine (0.1 mg/kg) and caffeine (20 mg/kg). Behavioral and physiological changes were evaluated by an assessment of physical signs, open field tests, elevated plus maze experiments, forced swimming tests, hot plate tests, and new-object-recognition tests. The changes in dopamine release in the prefrontal cortex (PFC) and ventral tegmental area (VTA) in the midbrain were analyzed using ELISA. The results showed that a combination of caffeine and nicotine could effectively relieve nicotine withdrawal syndrome, increase movement ability and pain thresholds, reduce anxiety and depression, enhance memory and cognitive ability, and increase the level of dopamine release in the PFC and VTA. Thus, caffeine combined with nicotine has potential as a stable and effective treatment option to help humans with smoking cessation.


Asunto(s)
Cafeína , Ratones Endogámicos C57BL , Nicotina , Síndrome de Abstinencia a Sustancias , Tabaquismo , Animales , Cafeína/farmacología , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Nicotina/farmacología , Masculino , Ratones , Tabaquismo/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Dopamina/metabolismo , Ansiedad/tratamiento farmacológico , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Modelos Animales de Enfermedad
2.
Transl Psychiatry ; 14(1): 391, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341817

RESUMEN

The consumption of alcohol and caffeine affects the lives of billions of individuals worldwide. Although recent evidence indicates that caffeine impairs the reinforcing properties of alcohol, a characterization of its effects on alcohol-stimulated mesolimbic dopamine (DA) function was lacking. Acting as the pro-drug of salsolinol, alcohol excites DA neurons in the posterior ventral tegmental area (pVTA) and increases DA release in the nucleus accumbens shell (AcbSh). Here we show that caffeine, via antagonistic activity on A2A adenosine receptors (A2AR), prevents alcohol-dependent activation of mesolimbic DA function as assessed, in-vivo, by brain microdialysis of AcbSh DA and, in-vitro, by electrophysiological recordings of pVTA DA neuronal firing. Accordingly, while the A1R antagonist DPCPX fails to prevent the effects of alcohol on DA function, both caffeine and the A2AR antagonist SCH 58261 prevent alcohol-dependent pVTA generation of salsolinol and increase in AcbSh DA in-vivo, as well as alcohol-dependent excitation of pVTA DA neurons in-vitro. However, caffeine also prevents direct salsolinol- and morphine-stimulated DA function, suggesting that it can exert these inhibitory effects also independently from affecting alcohol-induced salsolinol formation or bioavailability. Finally, untargeted metabolomics of the pVTA showcases that caffeine antagonizes alcohol-mediated effects on molecules (e.g. phosphatidylcholines, fatty amides, carnitines) involved in lipid signaling and energy metabolism, which could represent an additional salsolinol-independent mechanism of caffeine in impairing alcohol-mediated stimulation of mesolimbic DA transmission. In conclusion, the outcomes of this study strengthen the potential of caffeine, as well as of A2AR antagonists, for future development of preventive/therapeutic strategies for alcohol use disorder.


Asunto(s)
Cafeína , Dopamina , Neuronas Dopaminérgicas , Etanol , Núcleo Accumbens , Área Tegmental Ventral , Animales , Cafeína/farmacología , Dopamina/metabolismo , Etanol/farmacología , Masculino , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Ratas , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2A/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Antagonistas del Receptor de Adenosina A2/farmacología , Isoquinolinas
3.
Neurochem Res ; 49(11): 3131-3142, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39190121

RESUMEN

Emotional stress is a significant environmental risk factor for various mental health disabilities, such as anxiety. Electroacupuncture (EA) has been demonstrated to have pronounced anxiolytic effects. However, the neural mechanisms underlying these effects and their contribution to behavioral deficits remain poorly understood. Here, we addressed these issues using a classical mouse anxiety model induced by chronic restraint stress (CRS).Anxiety-like behaviors were evaluated with the open field test and elevated plus maze. Neuronal activation in various brain regions was marked using c-Fos, followed by calculations of interregional correlation to characterize a network that became functionally active following EA at the HT7 acupoint (EA-HT7). We selected the hub regions and further investigated their functions and connections in regulating anxiety-like behaviors by using a combination of chemogenetic manipulations and behavioral testing. CRS exposure induced anxiety-like behaviors. Interestingly, EA-HT7 mitigated these behavioral abnormalities. The c-Fos expression in 30 brain areas revealed a vital brain network for acupuncture responsiveness in naïve mice. Neural activity in the NAcSh (nucleus accumbens shell), BNST (bed nucleus of the stria terminalis), VMH (Ventromedial Hypothalamus), ARC (arcuate nucleus), dDG (dorsal dentate gyrus), and VTA (ventral tegmental area) was significantly altered following acupuncture. Notably, both c-Fos immunostaining and brain functional connectivity analysis revealed the significant activation of VTA following EA-HT7. Interestingly, blocking the VTA eliminated the anxiolytic effects of EA-HT7, whereas chemogenetic activation of the VTA replicated the therapeutic effects of EA-HT7. EA-HT7 has demonstrated benefits in treating anxiety and enhances brain functional connectivity. The VTA is functionally associated with the anxiolytic effects of EA-HT7.


Asunto(s)
Ansiedad , Electroacupuntura , Restricción Física , Estrés Psicológico , Área Tegmental Ventral , Animales , Electroacupuntura/métodos , Ansiedad/terapia , Ansiedad/psicología , Masculino , Área Tegmental Ventral/metabolismo , Estrés Psicológico/terapia , Estrés Psicológico/psicología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/metabolismo , Conducta Animal
4.
Sci Signal ; 17(848): eadk1822, 2024 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-39106321

RESUMEN

Deposition of amyloid-ß (Aß) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aß degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aß-degrading enzyme neprilysin and reduced the amount of Aß deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aß deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aß, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Dopamina , Neprilisina , Área Tegmental Ventral , Neprilisina/metabolismo , Neprilisina/genética , Animales , Dopamina/metabolismo , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Ratones , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Levodopa/farmacología , Encéfalo/metabolismo , Ratones Transgénicos , Modelos Animales de Enfermedad , Humanos , Proteolisis/efectos de los fármacos , Ratones Endogámicos C57BL , Corteza Prefrontal/metabolismo , Masculino
5.
Neuropharmacology ; 258: 110055, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38950692

RESUMEN

Sleep disturbances and persistent pain conditions are public health challenges worldwide. Although it is well-known that sleep deficit increases pain sensitivity, the underlying mechanisms remain elusive. We have recently demonstrated the involvement of nucleus accumbens (NAc) and anterior cingulate cortex (ACC) in the pronociceptive effect of sleep restriction. In this study, we found that sleep restriction increases c-Fos expression in NAc and ACC, suggesting hyperactivation of these regions during prolonged wakefulness in male Wistar rats. Blocking adenosine A2A receptors in the NAc or GABAA receptors in the ventral tegmental area (VTA), dorsal raphe nucleus (DRN), or locus coeruleus (LC) effectively mitigated the pronociceptive effect of sleep restriction. In contrast, the blockade of GABAA receptors in each of these nuclei only transiently reduced carrageenan-induced hyperalgesia. Pharmacological activation of dopamine D2, serotonin 5-HT1A and noradrenaline alpha-2 receptors within the ACC also prevented the pronociceptive effect of sleep restriction. While pharmacological inhibition of these same monoaminergic receptors in the ACC restored the pronociceptive effect which had been prevented by the GABAergic disinhibition of the of the VTA, DRN or LC. Overall, these findings suggest that the pronociceptive effect of sleep restriction relies on increased adenosinergic activity on NAc, heightened GABAergic activity in VTA, DRN, and LC, and reduced inhibitory monoaminergic activity on ACC. These findings advance our understanding of the interplay between sleep and pain, shedding light on potential NAc-brainstem-ACC mechanisms that could mediate increased pain sensitivity under conditions of sleep impairment.


Asunto(s)
Núcleo Accumbens , Ratas Wistar , Privación de Sueño , Área Tegmental Ventral , Animales , Masculino , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Ratas , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Receptor de Adenosina A2A/metabolismo , Hiperalgesia/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Giro del Cíngulo/metabolismo , Giro del Cíngulo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Tronco Encefálico/metabolismo , Tronco Encefálico/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/efectos de los fármacos , Carragenina , Receptores de GABA-A/metabolismo , Receptores de Dopamina D2/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología
6.
Obesity (Silver Spring) ; 32(8): 1448-1452, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38979671

RESUMEN

OBJECTIVE: This study aimed to determine a dopaminergic circuit required for diet-induced obesity in mice. METHODS: We created conditional deletion mutants for tyrosine hydroxylase (TH) using neurotensin receptor 1 (Ntsr1) Cre and other Cre drivers and measured feeding and body weight on standard and high-fat diets. We then used an adeno-associated virus to selectively restore TH to the ventral tegmental area (VTA) Ntsr1 neurons in conditional knockout (cKO) mice. RESULTS: Mice with cKO of Th using Vglut2-Cre, Cck-Cre, Calb1-Cre, and Bdnf-Cre were susceptible to obesity on a high-fat diet; however, Ntsr1-Cre Th cKO mice resisted weight gain on a high-fat diet and did not experience an increase in day eating unlike their wild-type littermate controls. Restoration of TH to the VTA Ntsr1 neurons of the Ntsr1-Cre Th cKO mice using an adeno-associated virus resulted in an increase in weight gain and day eating on a high-fat diet. CONCLUSIONS: Ntsr1-Cre Th cKO mice failed to increase day eating on a high-fat diet, offering a possible explanation for their resistance to diet-induced obesity. These results implicate VTA Ntsr1 dopamine neurons as promoting out-of-phase feeding behavior on a high-fat diet that could be an important contributor to diet-induced obesity in humans.


Asunto(s)
Dieta Alta en Grasa , Dopamina , Ratones Noqueados , Obesidad , Receptores de Neurotensina , Tirosina 3-Monooxigenasa , Área Tegmental Ventral , Aumento de Peso , Animales , Receptores de Neurotensina/metabolismo , Receptores de Neurotensina/genética , Obesidad/metabolismo , Obesidad/etiología , Ratones , Área Tegmental Ventral/metabolismo , Dopamina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Masculino , Neuronas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ratones Endogámicos C57BL , Dependovirus/genética , Peso Corporal
7.
Neuron ; 112(16): 2783-2798.e9, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-38959892

RESUMEN

The lateral septum (LS) is composed of heterogeneous cell types that are important for various motivated behaviors. However, the transcriptional profiles, spatial arrangement, function, and connectivity of these cell types have not been systematically studied. Using single-nucleus RNA sequencing, we delineated diverse genetically defined cell types in the LS that play distinct roles in reward processing. Notably, we found that estrogen receptor 1 (Esr1)-expressing neurons in the ventral LS (LSEsr1) are key drivers of reward seeking via projections to the ventral tegmental area, and these neurons play an essential role in methamphetamine (METH) reward and METH-seeking behavior. Extended exposure to METH increases the excitability of LSEsr1 neurons by upregulating hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, thereby contributing to METH-induced locomotor sensitization. These insights not only elucidate the intricate molecular, circuit, and functional architecture of the septal region in reward processing but also reveal a neural pathway critical for METH reward and behavioral sensitization.


Asunto(s)
Metanfetamina , Neuronas , Recompensa , Núcleos Septales , Animales , Ratones , Neuronas/fisiología , Neuronas/metabolismo , Metanfetamina/farmacología , Núcleos Septales/fisiología , Núcleos Septales/metabolismo , Masculino , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Vías Nerviosas/fisiología , Ratones Endogámicos C57BL , Comportamiento de Búsqueda de Drogas/fisiología
8.
Transl Psychiatry ; 14(1): 269, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956048

RESUMEN

Addiction is a complex behavioral disorder characterized by compulsive drug-seeking and drug use despite harmful consequences. The prefrontal cortex (PFC) plays a crucial role in cocaine addiction, involving decision-making, impulse control, memory, and emotional regulation. The PFC interacts with the brain's reward system, including the ventral tegmental area (VTA) and nucleus accumbens (NAc). The PFC also projects to the lateral habenula (LHb), a brain region critical for encoding negative reward and regulating the reward system. In the current study, we examined the role of PFC-LHb projections in regulating cocaine reward-related behaviors. We found that optogenetic stimulation of the PFC-LHb circuit during cocaine conditioning abolished cocaine preference without causing aversion. In addition, increased c-fos expression in LHb neurons was observed in animals that received optic stimulation during cocaine conditioning, supporting the circuit's involvement in cocaine preference regulation. Molecular analysis in animals that received optic stimulation revealed that cocaine-induced alterations in the expression of GluA1 subunit of AMPA receptor was normalized to saline levels in a region-specific manner. Moreover, GluA1 serine phosphorylation on S845 and S831 were differentially altered in LHb and VTA but not in the PFC. Together these findings highlight the critical role of the PFC-LHb circuit in controlling cocaine reward-related behaviors and shed light on the underlying mechanisms. Understanding this circuit's function may provide valuable insights into addiction and contribute to developing targeted treatments for substance use disorders.


Asunto(s)
Cocaína , Habénula , Neuronas , Optogenética , Corteza Prefrontal , Receptores AMPA , Recompensa , Animales , Corteza Prefrontal/metabolismo , Cocaína/farmacología , Masculino , Habénula/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Trastornos Relacionados con Cocaína/fisiopatología , Trastornos Relacionados con Cocaína/metabolismo , Vías Nerviosas , Ratas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Fosforilación , Área Tegmental Ventral/metabolismo , Conducta Animal
9.
Brain Behav Immun ; 120: 167-180, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38834156

RESUMEN

It is widely believed that the activation of the central dopamine (DA) system is crucial to the rewarding effects of methamphetamine (METH) and to the behavioral outcomes of METH use disorder. It was reported that METH exposure induced gasdermin D (GSDMD)-dependent pyroptosis in rats. The membrane pore formation caused by METH-induced pyroptosis may also contribute to the overflow of DA into the extracellular space and subsequently increase the DA levels in the brain. The present study firstly investigated whether the membrane pore information induced by GSDMD-dependent pyroptosis was associated with the increased DA levels in the ventral tegmental area (VAT) and nucleus accumbens (NAc) of rats self-administering METH and SY-SH5Y cells treated by METH. Subsequently, the effect of pore formation blockade or genetic inhibition of GSDMD on the reinforcing and motivational effect of METH was determined in rats, using the animal model of METH self-administration (SA). METH exposure significantly increased the activity of NLRP1/Cas-1/GSDMD pathway and the presence of pyroptosis, accompanied by the significantly increased DA levels in VTA and NAc. Moreover, intraperitoneal injections of disulfiram (DSF) or microinjection of rAAV-shGSDMD into VTA/NAc significantly reduced the reinforcing and motivational effect of METH, accompanied by the decreased level of DA in VTA and NAc. The results provided novel evidence that METH-induced pyroptosis could increase DA release in VTA and NAc via the NLRP1/Cas-1/GSDMD pathway. Additionally, membrane pores or GSDMD blockade could significantly reduce the reinforcing and motivational effect of METH. In conclusion, blocking GSDMD and membrane pore formation could be a promising potential target for the development of agents to treat METH use disorder.


Asunto(s)
Dopamina , Metanfetamina , Núcleo Accumbens , Proteínas de Unión a Fosfato , Piroptosis , Autoadministración , Área Tegmental Ventral , Animales , Metanfetamina/farmacología , Metanfetamina/administración & dosificación , Piroptosis/efectos de los fármacos , Masculino , Proteínas de Unión a Fosfato/metabolismo , Ratas , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Dopamina/metabolismo , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratas Sprague-Dawley , Humanos , Estimulantes del Sistema Nervioso Central/farmacología , Estimulantes del Sistema Nervioso Central/administración & dosificación , Gasderminas
10.
eNeuro ; 11(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38806231

RESUMEN

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.


Asunto(s)
Dopamina , Núcleo Accumbens , Receptores de Polipéptido Amiloide de Islotes Pancreáticos , Área Tegmental Ventral , Animales , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Masculino , Dopamina/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Receptores de Polipéptido Amiloide de Islotes Pancreáticos/metabolismo , Ratas Sprague-Dawley , Grasas de la Dieta/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Agonistas de los Receptores de Amilina/farmacología , Ratas , Sacarosa/administración & dosificación , Sacarosa/farmacología
11.
Int J Mol Sci ; 25(10)2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38791298

RESUMEN

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.


Asunto(s)
Núcleo Accumbens , Área Tegmental Ventral , Animales , Ratas , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Masculino , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/etiología , PPAR gamma/metabolismo , PPAR gamma/genética , Transducción de Señal/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Humo/efectos adversos , Nicotina/efectos adversos , Ratas Wistar , Nicotiana/efectos adversos , Tabaquismo/metabolismo , Fosforilación/efectos de los fármacos
12.
Int J Eat Disord ; 57(7): 1433-1446, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38650547

RESUMEN

OBJECTIVE: Binge-eating disorder is an eating disorder characterized by recurrent binge-eating episodes, during which individuals consume excessive amounts of highly palatable food (HPF) in a short time. This study investigates the intricate relationship between repeated binge-eating episode and the transcriptional regulation of two key genes, adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R), in selected brain regions of rats. METHOD: Binge-like eating behavior on HPF was induced through the combination of food restrictions and frustration stress (15 min exposure to HPF without access to it) in female rats, compared to control rats subjected to only restriction or only stress or none of these two conditions. After chronic binge-eating episodes, nucleic acids were extracted from different brain regions, and gene expression levels were assessed through real-time quantitative PCR. The methylation pattern on genes' promoters was investigated using pyrosequencing. RESULTS: The analysis revealed A2AAR upregulation in the amygdala and in the ventral tegmental area (VTA), and D2R downregulation in the nucleus accumbens in binge-eating rats. Concurrently, site-specific DNA methylation alterations at gene promoters were identified in the VTA for A2AAR and in the amygdala and caudate putamen for D2R. DISCUSSION: The alterations on A2AAR and D2R genes regulation highlight the significance of epigenetic mechanisms in the etiology of binge-eating behavior, and underscore the potential for targeted therapeutic interventions, to prevent the development of this maladaptive feeding behavior. These findings provide valuable insights for future research in the field of eating disorders. PUBLIC SIGNIFICANCE: Using an animal model with face, construct, and predictive validity, in which cycles of food restriction and frustration stress evoke binge-eating behavior, we highlight the significance of epigenetic mechanisms on adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R) genes regulation. They could represent new potential targets for the pharmacological management of eating disorders characterized by this maladaptive feeding behavior.


Asunto(s)
Trastorno por Atracón , Bulimia , Receptor de Adenosina A2A , Receptores de Dopamina D2 , Recompensa , Animales , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D2/genética , Femenino , Ratas , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Bulimia/metabolismo , Bulimia/genética , Trastorno por Atracón/genética , Trastorno por Atracón/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Metilación de ADN , Área Tegmental Ventral/metabolismo , Conducta Alimentaria , Núcleo Accumbens/metabolismo , Ratas Sprague-Dawley
13.
eNeuro ; 11(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38233142

RESUMEN

The medial habenula (MHb) has been identified as the limiting factor for nicotine intake and facilitating nicotine withdrawal. However, few studies have assessed MHb neuronal excitability in response to nicotine, and, currently, a gap in knowledge is present for finding behavioral correlates to neuronal excitability in the region. Moreover, no study to date has evaluated sex or nicotine dosage as factors of excitability in the MHb. Here, we utilized an e-vape self-administration (EVSA) model to determine differences between sexes with different nicotine dosages ± menthol. Following this paradigm, we employed patch-clamp electrophysiology to assess key metrics of MHb neuronal excitability in relation to behavioral endpoints. We observed female mice self-administered significantly more than males, regardless of dosage. We also observed a direct correlation between self-administration behavior and MHb excitability with low-dose nicotine + menthol in males. Conversely, a high dose of nicotine ± menthol yields an inverse correlation between excitability and self-administration behavior in males only. In addition, intrinsic excitability in the ventral tegmental area (VTA) does not track with the amount of nicotine self-administered. Rather, they correlate to the active/inactive discrimination of mice. Using fast-scan cyclic voltammetry, we also observed that dopamine release dynamics are linked to reinforcement-related behavior in males and motivation-related behaviors in females. These results point to a sex-specific difference in the activity of the MHb and VTA leading to distinct differences in self-administration behavior. His could lend evidence to clinical observations of smoking and nicotine-use behavior differing between males and females.


Asunto(s)
Habénula , Receptores Nicotínicos , Masculino , Femenino , Ratones , Animales , Nicotina/farmacología , Mentol/farmacología , Receptores Nicotínicos/metabolismo , Área Tegmental Ventral/metabolismo , Habénula/metabolismo
14.
Neurochem Res ; 49(1): 143-156, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37642894

RESUMEN

Several preclinical and clinical studies indicate that exposure to acute stress may decrease pain perception and increases pain tolerance. This phenomenon is called stress-induced analgesia (SIA). A variety of neurotransmitters, including dopamine, is involved in the SIA. Dopaminergic neurons in the mesolimbic circuits, originating from the ventral tegmental area (VTA), play a crucial role in various motivational, rewarding, and pain events. The present study aimed to investigate the modulatory role of VTA dopaminergic receptors in the antinociceptive responses evoked by forced swim stress (FSS) in a model of acute pain. One hundred-five adult male albino Wistar rats were subjected to stereotaxic surgery for implanting a unilateral cannula into the VTA. After one week of recovery, separate groups of animals were given different doses of SCH23390 and Sulpiride (0.25, 1, and 4 µg/0.3 µl) as D1- and D2-like receptor antagonists into the VTA, respectively. Then, the animals were exposed to FSS for a 6-min period, and the pain threshold was measured using the tail-flick test over a 60-min time set intervals. Results indicated that exposure to FSS produces a prominent antinociceptive response, diminishing by blocking both dopamine receptors in the VTA. Nonetheless, the effect of a D1-like dopamine receptor antagonist on FSS-induced analgesia was more prominent than that of a D2-like dopamine receptor antagonist. The results demonstrated that VTA dopaminergic receptors contribute to the pain process in stressful situations, and it might be provided a practical approach to designing new therapeutic agents for pain management.


Asunto(s)
Núcleo Accumbens , Área Tegmental Ventral , Ratas , Masculino , Animales , Área Tegmental Ventral/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D1/metabolismo , Antagonistas de Dopamina/farmacología , Ratas Wistar , Dolor/tratamiento farmacológico , Analgésicos/farmacología
15.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38003493

RESUMEN

Dopamine (DA)'s relationship with addiction is complex, and the related pathways in the mesocorticolimbic system are used to deliver DA, regulating both behavioral and perceptual actions. Specifically, the mesolimbic pathway connecting the ventral tegmental area (VTA) and the nucleus accumbens (NAc) is crucial in regulating memory, emotion, motivation, and behavior due to its responsibility to modulate dopamine. To better investigate the relationship between DA and addiction, more advanced mapping methods are necessary to monitor its production and propagation accurately and efficiently. In this study, we incorporate dLight1.2 adeno-associated virus (AAV) into our latest CMOS (complementary metal-oxide semiconductor) imaging platform to investigate the effects of two pharmacological substances, morphine and cocaine, in the NAc using adult mice. By implanting our self-fabricated CMOS imaging device into the deep brain, fluorescence imaging of the NAc using the dLight1.2 AAV allows for the visualization of DA molecules delivered from the VTA in real time. Our results suggest that changes in extracellular DA can be observed with this adapted system, showing potential for new applications and methods for approaching addiction studies. Additionally, we can identify the unique characteristic trend of DA release for both morphine and cocaine, further validating the underlying biochemical mechanisms used to modulate dopaminergic activation.


Asunto(s)
Cocaína , Ratones , Animales , Dopamina/metabolismo , Morfina/farmacología , Morfina/metabolismo , Núcleo Accumbens/metabolismo , Área Tegmental Ventral/metabolismo
16.
Nat Med ; 29(8): 2030-2040, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37580533

RESUMEN

Alcohol use disorder (AUD) exacts enormous personal, social and economic costs globally. Return to alcohol use in treatment-seeking patients with AUD is common, engendered by a cycle of repeated abstinence-relapse episodes even with use of currently available pharmacotherapies. Repeated ethanol use induces dopaminergic signaling neuroadaptations in ventral tegmental area (VTA) neurons of the mesolimbic reward pathway, and sustained dysfunction of reward circuitry is associated with return to drinking behavior. We tested this hypothesis by infusing adeno-associated virus serotype 2 vector encoding human glial-derived neurotrophic factor (AAV2-hGDNF), a growth factor that enhances dopaminergic neuron function, into the VTA of four male rhesus monkeys, with another four receiving vehicle, following induction of chronic alcohol drinking. GDNF expression ablated the return to alcohol drinking behavior over a 12-month period of repeated abstinence-alcohol reintroduction challenges. This behavioral change was accompanied by neurophysiological modulations to dopamine signaling in the nucleus accumbens that countered the hypodopaminergic signaling state associated with chronic alcohol use, indicative of a therapeutic modulation of limbic circuits countering the effects of alcohol. These preclinical findings suggest gene therapy targeting relapse prevention may be a potential therapeutic strategy for AUD.


Asunto(s)
Alcoholismo , Animales , Masculino , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/terapia , Alcoholismo/tratamiento farmacológico , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Etanol/metabolismo , Etanol/farmacología , Etanol/uso terapéutico , Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Núcleo Accumbens/metabolismo , Primates/genética , Área Tegmental Ventral/metabolismo
17.
eNeuro ; 10(5)2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37193602

RESUMEN

Mesolimbic nicotinic acetylcholine receptor (nAChRs) activation is necessary for nicotine reinforcement behavior, but it is unknown whether selective activation of nAChRs in the dopamine (DA) reward pathway is sufficient to support nicotine reinforcement. In this study, we tested the hypothesis that activation of ß2-containing (ß2*) nAChRs on VTA neurons is sufficient for intravenous nicotine self-administration (SA). We expressed ß2 nAChR subunits with enhanced sensitivity to nicotine (referred to as ß2Leu9'Ser) in the VTA of male Sprague Dawley (SD) rats, enabling very low concentrations of nicotine to selectively activate ß2* nAChRs on transduced neurons. Rats expressing ß2Leu9'Ser subunits acquired nicotine SA at 1.5 µg/kg/infusion, a dose too low to support acquisition in control rats. Saline substitution extinguished responding for 1.5 µg/kg/inf, verifying that this dose was reinforcing. ß2Leu9'Ser nAChRs also supported acquisition at the typical training dose in rats (30 µg/kg/inf) and reducing the dose to 1.5 µg/kg/inf caused a significant increase in the rate of nicotine SA. Viral expression of ß2Leu9'Ser subunits only in VTA DA neurons (via TH-Cre rats) also enabled acquisition of nicotine SA at 1.5 µg/kg/inf, and saline substitution significantly attenuated responding. Next, we examined electrically-evoked DA release in slices from ß2Leu9'Ser rats with a history of nicotine SA. Single-pulse evoked DA release and DA uptake rate were reduced in ß2Leu9'Ser NAc slices, but relative increases in DA following a train of stimuli were preserved. These results are the first to report that ß2* nAChR activation on VTA neurons is sufficient for nicotine reinforcement in rats.


Asunto(s)
Nicotina , Receptores Nicotínicos , Ratas , Masculino , Animales , Nicotina/farmacología , Nicotina/metabolismo , Agonistas Nicotínicos/farmacología , Área Tegmental Ventral/metabolismo , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Neuronas Dopaminérgicas/metabolismo
18.
Anat Sci Int ; 98(4): 580-592, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37160827

RESUMEN

Midbrain dopaminergic (DAergic) regions including ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) are involved in diverse brain functions. Previous studies demonstrated that the VTA/SNc to nucleus accumbens (NAc) pathway is critical in reward and motivation. Moreover, DAergic innervations within the insular cortex (IC) are reported to play important roles in pain regulation. To investigate whether VTA/SNc sends collateral projections to NAc and IC, we injected retrograde tracer Fluoro-Gold (FG) into the NAc and Fluorescent retrograde tracer beads (RetroBeads) into the ipsilateral IC in rats. Then, to detect whether collateral projection neurons participate in neuropathic pain, parts of the rats received the spare nerve injury (SNI) surgery. The immunofluorescence staining results showed that FG, RetroBeads, and FG/RetroBeads double-labeled neurons were distributed in the VTA/SNc bilaterally with an ipsilateral predominance. The proportion of FG/RetroBeads double-labeled neurons to the total number of FG and RetroBeads-labeled neurons was 16.7% and 30.3%, respectively. About 90.3% of FG/RetroBeads double-labeled neurons showed DAergic neuron marker tyrosine hydroxylase (TH)-immunoreactive (IR), whereas, only 7.5% exhibited a subset of GABAergic inhibitory projection neuron marker parvalbumin (PV)-IR. One week after SNI, about 53.1% and 33.6% of FG- and RetroBeads-labeled neurons were FG/Fos- and RetroBeads/Fos-IR neurons, respectively. Finally, about 35.9% of the FG/RetroBeads double-labeled neurons showed Fos-IR. The present study indicates that parts of DAergic and PV-IR GABAergic neurons in the VTA/SNc send collateral projections to both NAc and IC, which are activated under SNI-induced neuropathic pain, and probably contribute to the regulation of nociception.


Asunto(s)
Neuralgia , Área Tegmental Ventral , Ratas , Animales , Área Tegmental Ventral/metabolismo , Núcleo Accumbens/metabolismo , Porción Compacta de la Sustancia Negra/metabolismo , Corteza Insular , Sustancia Negra , Dopamina/metabolismo , Neuralgia/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
19.
Neuropharmacology ; 235: 109547, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37116611

RESUMEN

Nicotine is the principal psychoactive component in tobacco that drives addiction through its action on neuronal nicotinic acetylcholine receptors (nAChR). The nicotinic receptor gene CHRNA5, which encodes the α5 subunit, is associated with nicotine use and dependence. In humans, the CHRNA5 missense variant rs16969968 (G > A) is associated with increased risk for nicotine dependence and other smoking-related phenotypes. In rodents, α5-containing nAChRs in dopamine (DA) neurons within the ventral tegmental area (VTA) powerfully modulate nicotine reward and reinforcement. Although the neuroadaptations caused by long-term nicotine exposure are being actively delineated at both the synaptic and behavioral levels, the contribution of α5-containing nAChRs to the cellular adaptations associated with long-term nicotine exposure remain largely unknown. To gain insight into the mechanisms behind the influence of α5-containing nAChRs and the rs16969968 polymorphism on nicotine use and dependence, we used electrophysiological approaches to examine changes in nAChR function arising in VTA neurons during chronic nicotine exposure and multiple stages of nicotine withdrawal. Our results demonstrate that CHRNA5 mutation leads to profound changes in VTA nAChR function at baseline, during chronic nicotine exposure, and during short-term and prolonged withdrawal. Whereas nAChR function was suppressed in DA neurons from WT mice undergoing withdrawal relative to drug-naïve or nicotine-drinking mice, α5-null mice exhibited an increase in nAChR function during nicotine exposure that persisted throughout 5-10 weeks of withdrawal. Re-expressing the hypofunctional rs16969968 CHRNA5 variant in α5-null VTA DA neurons did not rescue the phenotype, with α5-SNP neurons displaying a similar increased response to ACh during nicotine exposure and early stages of withdrawal. These results demonstrate the importance of VTA α5-nAChRs in the response to nicotine and implicate them in the time course of withdrawal.


Asunto(s)
Nicotina , Receptores Nicotínicos , Humanos , Ratones , Animales , Nicotina/farmacología , Neuronas Dopaminérgicas/metabolismo , Área Tegmental Ventral/metabolismo , Receptores Nicotínicos/metabolismo , Fumar , Ratones Noqueados , Proteínas del Tejido Nervioso/genética
20.
Sci Rep ; 13(1): 4911, 2023 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-36966190

RESUMEN

Brain growth occurs during the first 2 weeks of postnatal development in rats. This developmental period is equivalent to the third trimester of human gestation. Dendritic arborization, axonal growth, and gliogenesis are observed along with a strong maturation of neurotransmission during this critical development period. Furthermore, nicotine exposure during early development causes deficiencies in sensory and cognitive processing in adults. In this study, we further investigated the gene expression of neuron groups and the influence of perinatal nicotine exposure on gene expressions of neurons within the sub-regions of the ventral tegmental area (VTA) in 1 week, 2 week and 3-week-old rat pups. We exposed pregnant rats to nicotine perinatally on gestational day 7 through postnatal day 14. Pups are exposed to nicotine during pregnancy and through breastfeeding to investigate its effect in rat pups during early neuronal development. Real time PCR was used to find the relative expressions of gamma-aminobutyric acid (GABA), dopamine, and glutamate neuron markers within the three sub-regions of the VTA including the parabrachial pigmented nucleus (PBP), parainterfascicular (PIF), and paranigral nucleus (PN). Our results indicated that during early maturation, the dopamine marker tyrosine hydroxylase (TH) showed a consistently increased significance in PN sub-region compared to PIF and PBP. These results suggest that following perinatal nicotine exposure, VTA dopamine neurons, especially within the PN sub-region, are significantly excited starting from birth.


Asunto(s)
Nicotina , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Ratas , Animales , Nicotina/efectos adversos , Nicotina/metabolismo , Área Tegmental Ventral/metabolismo , Transcriptoma , Dopamina/metabolismo , Ratas Sprague-Dawley , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Neuronas Dopaminérgicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA