Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.628
Filtrar
1.
Transl Psychiatry ; 14(1): 277, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965230

RESUMEN

The mechanisms contributing to alcohol use disorder (AUD) are complex and the orexigenic peptide ghrelin, which enhances alcohol reward, is implied as a crucial modulator. The major proportion of circulating ghrelin is however the non-octanoylated form of ghrelin, des-acyl ghrelin (DAG), whose role in reward processes is unknown. As recent studies show that DAG decreases food intake, we hypothesize that DAG attenuates alcohol-related responses in animal models. Acute and repeated DAG treatment dose-dependently decreased alcohol drinking in male and female rats. In these alcohol-consuming male rats, repeated DAG treatment causes higher levels of dopamine metabolites in the ventral tegmental area, an area central to reward processing. The role of DAG in reward processing is further supported as DAG prevents alcohol-induced locomotor stimulation, reward in the conditioned place preference paradigm, and dopamine release in the nucleus accumbens in male rodents. On the contrary, DAG does not alter the memory of alcohol reward or affect neurotransmission in the hippocampus, an area central to memory. Further, circulating DAG levels are positively correlated with alcohol drinking in female but not male rats. Studies were conducted in attempts to identify tentative targets of DAG, which currently are unknown. Data from these recombinant cell system revealed that DAG does not bind to either of the monoamine transporters, 5HT2A, CB1, or µ-opioid receptors. Collectively, our data show that DAG attenuates alcohol-related responses in rodents, an effect opposite to that of ghrelin, and contributes towards a deeper insight into behaviors regulated by the ghrelinergic signaling pathway.


Asunto(s)
Consumo de Bebidas Alcohólicas , Dopamina , Ghrelina , Núcleo Accumbens , Recompensa , Área Tegmental Ventral , Animales , Ghrelina/farmacología , Ghrelina/metabolismo , Masculino , Ratas , Femenino , Dopamina/metabolismo , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Etanol/farmacología , Etanol/administración & dosificación , Humanos , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratas Sprague-Dawley
2.
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
3.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 402-408, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38953264

RESUMEN

There are mutual neural projections between the ventral tegmental area (VTA) and the medial prefrontal cortex (mPFC),which form a circuit.Recent studies have shown that this circuit is vital in regulating arousal from sleep and general anesthesia.This paper introduces the anatomical structures of VTA and mPFC and the roles of various neurons and projection pathways in the regulation of arousal,aiming to provide new ideas for further research on the mechanism of arousal from sleep and general anesthesia.


Asunto(s)
Nivel de Alerta , Corteza Prefrontal , Área Tegmental Ventral , Corteza Prefrontal/fisiología , Área Tegmental Ventral/fisiología , Nivel de Alerta/fisiología , Humanos , Animales , Vías Nerviosas/fisiología
4.
Int J Mol Sci ; 25(12)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38928178

RESUMEN

Physiology and behavior are structured temporally to anticipate daily cycles of light and dark, ensuring fitness and survival. Neuromodulatory systems in the brain-including those involving serotonin and dopamine-exhibit daily oscillations in neural activity and help shape circadian rhythms. Disrupted neuromodulation can cause circadian abnormalities that are thought to underlie several neuropsychiatric disorders, including bipolar mania and schizophrenia, for which a mechanistic understanding is still lacking. Here, we show that genetically depleting serotonin in Tph2 knockout mice promotes manic-like behaviors and disrupts daily oscillations of the dopamine biosynthetic enzyme tyrosine hydroxylase (TH) in midbrain dopaminergic nuclei. Specifically, while TH mRNA and protein levels in the Substantia Nigra (SN) and Ventral Tegmental Area (VTA) of wild-type mice doubled between the light and dark phase, TH levels were high throughout the day in Tph2 knockout mice, suggesting a hyperdopaminergic state. Analysis of TH expression in striatal terminal fields also showed blunted rhythms. Additionally, we found low abundance and blunted rhythmicity of the neuropeptide cholecystokinin (Cck) in the VTA of knockout mice, a neuropeptide whose downregulation has been implicated in manic-like states in both rodents and humans. Altogether, our results point to a previously unappreciated serotonergic control of circadian dopamine signaling and propose serotonergic dysfunction as an upstream mechanism underlying dopaminergic deregulation and ultimately maladaptive behaviors.


Asunto(s)
Ritmo Circadiano , Dopamina , Ratones Noqueados , Serotonina , Triptófano Hidroxilasa , Tirosina 3-Monooxigenasa , Área Tegmental Ventral , Animales , Serotonina/metabolismo , Ratones , Ritmo Circadiano/fisiología , Dopamina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Tirosina 3-Monooxigenasa/genética , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/deficiencia , Área Tegmental Ventral/metabolismo , Colecistoquinina/metabolismo , Colecistoquinina/genética , Neuronas Dopaminérgicas/metabolismo , Masculino , Sustancia Negra/metabolismo , Ratones Endogámicos C57BL , Trastorno Bipolar/metabolismo , Trastorno Bipolar/genética
5.
Behav Neurosci ; 138(3): 164-177, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38934920

RESUMEN

A growing body of literature indicates that mediated learning techniques have specific utility for tapping into reality testing in animal models of neuropsychiatric illness. In particular, recent work has shown that animal models that recapitulate various endophenotypes of schizophrenia are particularly vulnerable to impairments in reality testing when undergoing mediated learning. Multiple studies have indicated that these effects are dopamine receptor 2-dependent and correlated with aberrant insular cortex (IC) activity. However, until now, the connection between dopamine and the IC had not been investigated. Here, we utilized a novel intersectional approach to label mesencephalic dopamine cells that specifically project to the insular cortex in both wild-type controls and transgenic mice expressing the dominant-negative form of the Disrupted-in-Schizophrenia-1 (DISC-1) gene. Using these techniques, we identified a population of cells that project from the ventral tegmental area (VTA) to the IC. Afterward, we conducted multiple studies to test the necessity of this circuit in behaviors ranging from gustatory detection to the maintenance of effort and, finally, mediated performance. Our results indicate that perturbations of the DISC-1 genetic locus lead to a reduction in the number of cells in the VTA → IC circuit. Behaviorally, VTA → IC circuitry does not influence gustatory detection or motivation to acquire sucrose reward; however, inactivation of this circuit differentially suppresses Pavlovian approach behavior in wild-type and DISC-1 transgenic mice during mediated performance testing. Moreover, under these testing conditions, inactivation of this circuit predisposes wild-type (but not DISC-1) mice to display impaired reality testing. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Asunto(s)
Neuronas Dopaminérgicas , Corteza Insular , Ratones Transgénicos , Animales , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/metabolismo , Ratones , Corteza Insular/fisiología , Masculino , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Recompensa , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Mesencéfalo/metabolismo , Mesencéfalo/fisiología , Esquizofrenia/fisiopatología
6.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892125

RESUMEN

A total of 3102 neurons were recorded before and following acute and chronic methylphenidate (MPD) administration. Acute MPD exposure elicits mainly increases in neuronal and behavioral activity in dose-response characteristics. The response to chronic MPD exposure, as compared to acute 0.6, 2.5, or 10.0 mg/kg MPD administration, elicits electrophysiological and behavioral sensitization in some animals and electrophysiological and behavioral tolerance in others when the neuronal recording evaluations were performed based on the animals' behavioral responses, or amount of locomotor activity, to chronic MPD exposure. The majority of neurons recorded from those expressing behavioral sensitization responded to chronic MPD with further increases in firing rate as compared to the initial MPD responses. The majority of neurons recorded from animals expressing behavioral tolerance responded to chronic MPD with decreases in their firing rate as compared to the initial MPD exposures. Each of the six brain areas studied-the ventral tegmental area, locus coeruleus, dorsal raphe, nucleus accumbens, prefrontal cortex, and caudate nucleus (VTA, LC, DR, NAc, PFC, and CN)-responds significantly (p < 0.001) differently to MPD, suggesting that each one of the above brain areas exhibits different roles in the response to MPD. Moreover, this study demonstrates that it is essential to evaluate neuronal activity responses to psychostimulants based on the animals' behavioral responses to acute and chronic effects of the drug from several brain areas simultaneously to obtain accurate information on each area's role in response to the drug.


Asunto(s)
Conducta Animal , Núcleo Caudado , Metilfenidato , Neuronas , Núcleo Accumbens , Corteza Prefrontal , Área Tegmental Ventral , Animales , Metilfenidato/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/metabolismo , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/fisiología , Núcleo Caudado/metabolismo , Masculino , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Conducta Animal/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/fisiología , Ratas Sprague-Dawley , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/fisiología , Núcleo Dorsal del Rafe/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología
7.
Nat Commun ; 15(1): 4947, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858350

RESUMEN

The potential brain mechanism underlying resilience to socially transferred allodynia remains unknown. Here, we utilize a well-established socially transferred allodynia paradigm to segregate male mice into pain-susceptible and pain-resilient subgroups. Brain screening results show that ventral tegmental area glutamatergic neurons are selectively activated in pain-resilient mice as compared to control and pain-susceptible mice. Chemogenetic manipulations demonstrate that activation and inhibition of ventral tegmental area glutamatergic neurons bi-directionally regulate resilience to socially transferred allodynia. Moreover, ventral tegmental area glutamatergic neurons that project specifically to the nucleus accumbens shell and lateral habenula regulate the development and maintenance of the pain-resilient phenotype, respectively. Together, we establish an approach to explore individual variations in pain response and identify ventral tegmental area glutamatergic neurons and related downstream circuits as critical targets for resilience to socially transferred allodynia and the development of conceptually innovative analgesics.


Asunto(s)
Ácido Glutámico , Hiperalgesia , Neuronas , Núcleo Accumbens , Área Tegmental Ventral , Animales , Masculino , Hiperalgesia/fisiopatología , Área Tegmental Ventral/fisiopatología , Ratones , Ácido Glutámico/metabolismo , Núcleo Accumbens/fisiopatología , Neuronas/metabolismo , Mesencéfalo , Ratones Endogámicos C57BL , Resiliencia Psicológica , Habénula , Modelos Animales de Enfermedad
8.
Nat Commun ; 15(1): 5353, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918403

RESUMEN

Nociceptin/orphanin-FQ (N/OFQ) is a recently appreciated critical opioid peptide with key regulatory functions in several central behavioral processes including motivation, stress, feeding, and sleep. The functional relevance of N/OFQ action in the mammalian brain remains unclear due to a lack of high-resolution approaches to detect this neuropeptide with appropriate spatial and temporal resolution. Here we develop and characterize NOPLight, a genetically encoded sensor that sensitively reports changes in endogenous N/OFQ release. We characterized the affinity, pharmacological profile, spectral properties, kinetics, ligand selectivity, and potential interaction with intracellular signal transducers of NOPLight in vitro. Its functionality was established in acute brain slices by exogeneous N/OFQ application and chemogenetic induction of endogenous N/OFQ release from PNOC neurons. In vivo studies with fibre photometry enabled direct recording of NOPLight binding to exogenous N/OFQ receptor ligands, as well as detection of endogenous N/OFQ release within the paranigral ventral tegmental area (pnVTA) during natural behaviors and chemogenetic activation of PNOC neurons. In summary, we show here that NOPLight can be used to detect N/OFQ opioid peptide signal dynamics in tissue and freely behaving animals.


Asunto(s)
Neuronas , Nociceptina , Péptidos Opioides , Receptores Opioides , Animales , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Receptores Opioides/genética , Neuronas/metabolismo , Humanos , Ratones , Masculino , Área Tegmental Ventral/metabolismo , Receptor de Nociceptina , Células HEK293 , Encéfalo/metabolismo , Ratones Endogámicos C57BL , Ligandos , Técnicas Biosensibles/métodos
9.
Nature ; 630(8017): 677-685, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38839962

RESUMEN

All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.


Asunto(s)
Analgésicos Opioides , Vaina de Mielina , Vías Nerviosas , Plasticidad Neuronal , Recompensa , Área Tegmental Ventral , Animales , Femenino , Masculino , Ratones , Analgésicos Opioides/farmacología , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Ratones Endogámicos C57BL , Morfina/farmacología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiología , Núcleo Accumbens/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Optogenética , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Linaje de la Célula
10.
Elife ; 132024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38865180

RESUMEN

A social memory pathway connecting the ventral hippocampus, the lateral septum and the ventral tegmental area helps to regulate how mice react to unknown individuals.


Asunto(s)
Conducta Social , Animales , Ratones , Área Tegmental Ventral/fisiología , Hipocampo/fisiología , Memoria/fisiología
11.
Nat Commun ; 15(1): 4233, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762463

RESUMEN

The ventral pallidum (VP) contains GABA and glutamate neurons projecting to ventral tegmental area (VTA) whose stimulation drives approach and avoidance, respectively. Yet little is known about the mechanisms by which VP cell types shape VTA activity and drive behavior. Here, we found that both VP GABA and glutamate neurons were activated during approach to reward or by delivery of an aversive stimulus. Stimulation of VP GABA neurons inhibited VTA GABA, but activated dopamine and glutamate neurons. Remarkably, stimulation-evoked activation was behavior-contingent such that VTA recruitment was inhibited when evoked by the subject's own action. Conversely, VP glutamate neurons activated VTA GABA, as well as dopamine and glutamate neurons, despite driving aversion. However, VP glutamate neurons evoked dopamine in aversion-associated ventromedial nucleus accumbens (NAc), but reduced dopamine release in reward-associated dorsomedial NAc. These findings show how heterogeneous VP projections to VTA can be engaged to shape approach and avoidance behaviors.


Asunto(s)
Reacción de Prevención , Prosencéfalo Basal , Neuronas GABAérgicas , Ácido Glutámico , Recompensa , Área Tegmental Ventral , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/citología , Animales , Ácido Glutámico/metabolismo , Prosencéfalo Basal/metabolismo , Prosencéfalo Basal/fisiología , Masculino , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Reacción de Prevención/fisiología , Ratones , Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Ácido gamma-Aminobutírico/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Ratones Endogámicos C57BL , Conducta Animal/fisiología
12.
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
13.
Nat Commun ; 15(1): 4152, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755120

RESUMEN

Serotonin is a neuromodulator that affects multiple behavioral and cognitive functions. Nonetheless, how serotonin causes such a variety of effects via brain-wide projections and various receptors remains unclear. Here we measured brain-wide responses to optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus (DRN) of the male mouse brain using functional MRI with an 11.7 T scanner and a cryoprobe. Transient activation of DRN serotonin neurons caused brain-wide activation, including the medial prefrontal cortex, the striatum, and the ventral tegmental area. The same stimulation under anesthesia with isoflurane decreased brain-wide activation, including the hippocampal complex. These brain-wide response patterns can be explained by DRN serotonergic projection topography and serotonin receptor expression profiles, with enhanced weights on 5-HT1 receptors. Together, these results provide insight into the DR serotonergic system, which is consistent with recent discoveries of its functions in adaptive behaviors.


Asunto(s)
Núcleo Dorsal del Rafe , Optogenética , Neuronas Serotoninérgicas , Serotonina , Animales , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Masculino , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Ratones , Serotonina/metabolismo , Imagen por Resonancia Magnética , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/fisiología , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Receptores de Serotonina/metabolismo , Receptores de Serotonina/genética
14.
Nat Commun ; 15(1): 4100, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773091

RESUMEN

In most models of neuronal plasticity and memory, dopamine is thought to promote the long-term maintenance of Long-Term Potentiation (LTP) underlying memory processes, but not the initiation of plasticity or new information storage. Here, we used optogenetic manipulation of midbrain dopamine neurons in male DAT::Cre mice, and discovered that stimulating the Schaffer collaterals - the glutamatergic axons connecting CA3 and CA1 regions - of the dorsal hippocampus concomitantly with midbrain dopamine terminals within a 200 millisecond time-window triggers LTP at glutamatergic synapses. Moreover, we showed that the stimulation of this dopaminergic pathway facilitates contextual learning in awake behaving mice, while its inhibition hinders it. Thus, activation of midbrain dopamine can operate as a teaching signal that triggers NeoHebbian LTP and promotes supervised learning.


Asunto(s)
Dopamina , Neuronas Dopaminérgicas , Hipocampo , Aprendizaje , Potenciación a Largo Plazo , Optogenética , Área Tegmental Ventral , Animales , Potenciación a Largo Plazo/fisiología , Área Tegmental Ventral/fisiología , Masculino , Dopamina/metabolismo , Ratones , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/metabolismo , Hipocampo/fisiología , Hipocampo/metabolismo , Aprendizaje/fisiología , Ratones Transgénicos , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/citología , Sinapsis/fisiología , Sinapsis/metabolismo , Ratones Endogámicos C57BL , Memoria/fisiología
15.
Alcohol Alcohol ; 59(4)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38742547

RESUMEN

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.


Asunto(s)
Consumo de Bebidas Alcohólicas , Neuronas , Quinina , Caracteres Sexuales , Animales , Quinina/farmacología , Femenino , Masculino , Ratones , Neuronas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Mesencéfalo/metabolismo , Mesencéfalo/efectos de los fármacos , Corteza Insular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Etanol/farmacología , Ácido Glutámico/metabolismo
16.
Behav Brain Res ; 468: 115040, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38723675

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Hidrógeno , Fármacos Neuroprotectores , Oxidopamina , Silicio , Animales , Fármacos Neuroprotectores/farmacología , Oxidopamina/farmacología , Ratones , Silicio/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Femenino , Hidrógeno/farmacología , Hidrógeno/administración & dosificación , Masculino , Síndromes de Neurotoxicidad/tratamiento farmacológico , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Ratones Endogámicos C57BL
17.
Neurobiol Learn Mem ; 212: 107930, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38692391

RESUMEN

Positive social comparative feedback is hypothesized to generate a dopamine response in the brain, similar to reward, by enhancing expectancies to support motor skill learning. However, no studies have utilized neuroimaging to examine this hypothesized dopaminergic mechanism. Therefore, the aim of this preliminary study was to investigate the effect of positive social comparative feedback on dopaminergic neural pathways measured by resting state connectivity. Thirty individuals practiced an implicit, motor sequence learning task and were assigned to groups that differed in feedback type. One group received feedback about their actual response time to complete the task (RT ONLY), while the other group received feedback about their response time with positive social comparison (RT + POS). Magnetic resonance imaging was acquired at the beginning and end of repetitive motor practice with feedback to measure practice-dependent changes in resting state brain connectivity. While both groups showed improvements in task performance and increases in performance expectancies, ventral tegmental area and the left nucleus accumbens (mesolimbic dopamine pathway) resting state connectivity increased in the RT + POS group but not in the RT ONLY group. Instead, the RT ONLY group showed increased connectivity between ventral tegmental area and primary motor cortex. Positive social comparative feedback during practice of a motor sequence task may induce a dopaminergic response in the brain along the mesolimbic pathway. However, given that absence of effects on expectancies and motor learning, more robust and individualized approaches may be needed to provide beneficial psychological and behavioral effects.


Asunto(s)
Imagen por Resonancia Magnética , Vías Nerviosas , Núcleo Accumbens , Área Tegmental Ventral , Humanos , Masculino , Femenino , Adulto Joven , Adulto , Área Tegmental Ventral/fisiología , Área Tegmental Ventral/diagnóstico por imagen , Vías Nerviosas/fisiología , Núcleo Accumbens/fisiología , Núcleo Accumbens/diagnóstico por imagen , Dopamina/metabolismo , Dopamina/fisiología , Retroalimentación Psicológica/fisiología , Corteza Motora/fisiología , Corteza Motora/diagnóstico por imagen , Encéfalo/fisiología , Encéfalo/diagnóstico por imagen , Destreza Motora/fisiología , Práctica Psicológica
18.
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
19.
Addict Biol ; 29(5): e13403, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38735880

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Fentanilo , Ratones Endogámicos C57BL , Área Tegmental Ventral , Animales , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Ratones , Fentanilo/farmacología , Masculino , Femenino , Analgésicos Opioides/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Autoadministración , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Relacionados con Opioides/genética
20.
Nature ; 630(8015): 141-148, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38778097

RESUMEN

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.


Asunto(s)
Fentanilo , Receptores Opioides mu , Refuerzo en Psicología , Animales , Femenino , Masculino , Ratones , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Núcleo Amigdalino Central/citología , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Fentanilo/farmacología , Ratones Endogámicos C57BL , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/patología , Optogenética , Receptores Opioides mu/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/patología , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...