Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 9(32): eadg8869, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37566654

RESUMO

Dopamine is broadly implicated in reinforcement learning, but how patterns of dopamine activity are generated is poorly resolved. Here, we demonstrate that two ion channels, Kv4.3 and BKCa1.1, regulate the pattern of dopamine neuron firing and dopamine release on different time scales to influence separate phases of reinforced behavior in mice. Inactivation of Kv4.3 in VTA dopamine neurons increases ex vivo pacemaker activity and excitability that is associated with increased in vivo firing rate and ramping dynamics before lever press in a learned instrumental paradigm. Loss of Kv4.3 enhances performance of the learned response and facilitates extinction. In contrast, loss of BKCa1.1 increases burst firing and phasic dopamine release that enhances learning of an instrumental response and enhances extinction burst lever pressing in early extinction that is associated with a greater change in activity between reinforced and unreinforced actions. These data demonstrate that disruption of intrinsic regulators of neuronal activity differentially affects dopamine dynamics during reinforcement and extinction learning.


Assuntos
Dopamina , Neurônios Dopaminérgicos , Camundongos , Animais , Reforço Psicológico , Aprendizagem , Canais Iônicos
2.
Nature ; 619(7969): 332-337, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37380765

RESUMO

Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.


Assuntos
Encéfalo , Vias Neurais , Neurotensina , Neurotransmissores , Transdução de Sinais , Encéfalo/citologia , Encéfalo/metabolismo , Cálcio/metabolismo , Sistemas CRISPR-Cas , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios GABAérgicos , Ácido gama-Aminobutírico/metabolismo , Edição de Genes , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Neurotensina/metabolismo , Neurotransmissores/metabolismo , Receptores de Neurotensina/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo
3.
bioRxiv ; 2023 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-37090565

RESUMO

In some individuals, drug-associated cues subsume potent control of behavior, such as the elicitation of drug craving1-3 and automatized drug use4. The intensity of this cue reactivity is highly predictive of relapse and other clinical outcomes in substance use disorders5,6. It has been postulated that this cue reactivity is driven by augmentation of dopamine release over the course of chronic drug use7. Here we carried out longitudinal recording and manipulation of cue-evoked dopamine signaling across phases of substance-use related behavior in rats. We observed a subset of individuals that exhibited increased cue reactivity and escalated drug consumption, two cardinal features of substance use disorders. In these individuals, cue-evoked phasic dopamine release underwent diametrically opposed changes in amplitude, determined by the context in which the cue is presented. Dopamine evoked by non-contingent cue presentation increased over drug use, producing greater cue reactivity; whereas dopamine evoked by contingent cue presentation decreased over drug use, producing escalation of drug consumption. Therefore, despite being in opposite directions, these dopamine trajectories each promote core symptoms of substance use disorders.

4.
Elife ; 122023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36927614

RESUMO

The axonal guidance cue netrin-1 serves a critical role in neural circuit development by promoting growth cone motility, axonal branching, and synaptogenesis. Within the adult mouse brain, expression of the gene encoding (Ntn1) is highly enriched in the ventral midbrain where it is expressed in both GABAergic and dopaminergic neurons, but its function in these cell types in the adult system remains largely unknown. To address this, we performed viral-mediated, cell-type specific CRISPR-Cas9 mutagenesis of Ntn1 in the ventral tegmental area (VTA) of adult mice. Ntn1 loss-of-function in either cell type resulted in a significant reduction in excitatory postsynaptic connectivity. In dopamine neurons, the reduced excitatory tone had a minimal phenotypic behavioral outcome; however, reduced glutamatergic tone on VTA GABA neurons induced behaviors associated with a hyperdopaminergic phenotype. Simultaneous loss of Ntn1 function in both cell types largely rescued the phenotype observed in the GABA-only mutagenesis. These findings demonstrate an important role for Ntn1 in maintaining excitatory connectivity in the adult midbrain and that a balance in this connectivity within two of the major cell types of the VTA is critical for the proper functioning of the mesolimbic system.


Assuntos
Ácido Glutâmico , Área Tegmentar Ventral , Camundongos , Animais , Ácido Glutâmico/metabolismo , Área Tegmentar Ventral/fisiologia , Netrina-1/metabolismo , Transdução de Sinais , Neurônios Dopaminérgicos/fisiologia
5.
J Neurophysiol ; 128(1): 62-72, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35788155

RESUMO

Ion channel complexes typically consist of both pore-forming subunits and auxiliary subunits that do not directly conduct current but can regulate trafficking or alter channel properties. Isolating the role of these auxiliary subunits in neurons has proved difficult due to a lack of specific pharmacological agents and the potential for developmental compensation in constitutive knockout models. Here, we use cell-type-specific viral-mediated CRISPR/Cas9 mutagenesis to target the potassium channel auxiliary subunit Kvß2 (Kcnab2) in dopamine neurons in the adult mouse brain. We find that mutagenesis of Kcnab2 reduces surface expression of Kv1.2, the primary Kv1 pore-forming subunit expressed in dopamine neurons, and shifts the voltage dependence of inactivation of potassium channel currents toward more hyperpolarized potentials. Loss of Kcnab2 broadens the action potential waveform in spontaneously firing dopamine neurons recorded in slice, reduces the afterhyperpolarization amplitude, and increases spike timing irregularity and excitability, all of which is consistent with a reduction in potassium channel current. Similar effects were observed with mutagenesis of the pore-forming subunit Kv1.2 (Kcna2). These results identify Kv1 currents as important contributors to dopamine neuron firing and demonstrate a role for Kvß2 subunits in regulating the trafficking and gating properties of these ion channels. Furthermore, they demonstrate the utility of CRISPR-mediated mutagenesis in the study of previously difficult to isolate ion channel subunits.NEW & NOTEWORTHY Here, we utilize CRISPR/Cas9-mediated mutagenesis in dopamine neurons in mice to target the gene encoding Kvß2, an auxiliary subunit that forms a part of Kv1 channel complexes. We find that the absence of Kvß2 alters action potential properties by reducing surface expression of pore-forming subunits and shifting the voltage dependence of channel inactivation. This work establishes a new function for Kvß2 subunits and Kv1 complexes in regulating dopamine neuron activity.


Assuntos
Neurônios Dopaminérgicos , Canais de Potássio , Animais , Neurônios Dopaminérgicos/metabolismo , Camundongos , Canais de Potássio/metabolismo , Superfamília Shaker de Canais de Potássio/genética
6.
Front Neural Circuits ; 16: 918839, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35860212

RESUMO

Neuropeptides play an important role in modulating mesolimbic system function. However, while synaptic inputs to the ventral tegmental area (VTA) have been extensively mapped, the sources of many neuropeptides are not well resolved. Here, we mapped the anatomical locations of three neuropeptide inputs to the VTA: neurotensin (NTS), corticotrophin releasing factor (CRF), and neurokinin B (NkB). Among numerous labeled inputs we identified the bed nucleus of the stria terminalis (BNST) as a major source of all three peptides, containing similar numbers of NTS, CRF, and NkB VTA projection neurons. Approximately 50% of BNST to VTA inputs co-expressed two or more of the peptides examined. Consistent with this expression pattern, analysis of calcium dynamics in the terminals of these inputs in the VTA revealed both common and distinct patterns of activation during appetitive and aversive conditioning. These data demonstrate additional diversification of the mesolimbic dopamine system through partially overlapping neuropeptidergic inputs.


Assuntos
Núcleos Septais , Área Tegmentar Ventral , Hormônio Liberador da Corticotropina/metabolismo , Neurônios/metabolismo , Recompensa , Núcleos Septais/metabolismo , Área Tegmentar Ventral/fisiologia
7.
Curr Biol ; 31(19): 4388-4396.e5, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34388372

RESUMO

Discrimination between predictive and non-predictive threat stimuli decreases as threat intensity increases. The central mechanisms that mediate the transition from discriminatory to generalized threat responding remain poorly resolved. Here, we identify the stress- and dysphoria-associated kappa opioid receptor (KOR) and its ligand dynorphin (Dyn), acting in the ventral tegmental area (VTA), as a key substrate for regulating threat generalization. We identify several dynorphinergic inputs to the VTA and demonstrate that projections from the bed nucleus of the stria terminalis (BNST) and dorsal raphe nucleus (DRN) both contribute to anxiety-like behavior but differentially affect threat generalization. These data demonstrate that conditioned threat discrimination has an inverted "U" relationship with threat intensity and establish a role for KOR/Dyn signaling in the midbrain for promoting threat generalization.


Assuntos
Dinorfinas , Núcleos Septais , Núcleo Dorsal da Rafe , Receptores Opioides kappa/metabolismo , Área Tegmentar Ventral/metabolismo
8.
Nat Neurosci ; 23(8): 968-980, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32541962

RESUMO

The ventral tegmental area (VTA) is important for reward processing and motivation. The anatomic organization of neurotransmitter-specific inputs to the VTA remains poorly resolved. In the present study, we mapped the major neurotransmitter projections to the VTA through cell-type-specific retrograde and anterograde tracing. We found that glutamatergic inputs arose from a variety of sources and displayed some connectivity biases toward specific VTA cell types. The sources of GABAergic projections were more widespread, displayed a high degree of differential innervation of subregions in the VTA and were largely biased toward synaptic contact with local GABA neurons. Inactivation of GABA release from the two major sources, locally derived versus distally derived, revealed distinct roles for these projections in behavioral regulation. Optogenetic manipulation of individual distal GABAergic inputs also revealed differential behavioral effects. These results demonstrate that GABAergic projections to the VTA are a major contributor to the regulation and diversification of the structure.


Assuntos
Neurônios GABAérgicos/metabolismo , Transmissão Sináptica/fisiologia , Área Tegmentar Ventral/metabolismo , Animais , Condicionamento Clássico/fisiologia , Condicionamento Operante/fisiologia , Neurônios Dopaminérgicos/fisiologia , Medo/fisiologia , Feminino , Masculino , Camundongos , Atividade Motora/fisiologia , Vias Neurais/metabolismo , Optogenética , Recompensa , Autoestimulação
9.
Cell Rep ; 30(12): 4303-4316.e6, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209486

RESUMO

Mice engineered for conditional, cell type-specific gene inactivation have dominated the field of mouse genetics because of the high efficiency of Cre-loxP-mediated recombination. Recent advances in CRISPR/Cas9 technologies have provided alternatives for rapid gene mutagenesis for loss-of-function (LOF) analysis. Whether these strategies can be streamlined for rapid genetic analysis with the efficiencies comparable with those of conventional genetic approaches has yet to be established. We show that a single adeno-associated viral (AAV) vector containing a recombinase-dependent Staphylococcus aureus Cas9 (SaCas9) and a single guide RNA (sgRNA) are as efficient as conventional conditional gene knockout and can be adapted for use in either Cre- or Flp-driver mouse lines. The efficacy of this approach is demonstrated for the analysis of GABAergic, glutamatergic, and monoaminergic neurotransmission. Using this strategy, we reveal insight into the role of GABAergic regulation of midbrain GABA-producing neurons in psychomotor activation.


Assuntos
Envelhecimento/genética , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Vetores Genéticos/metabolismo , Mutagênese/genética , Sistema Nervoso/metabolismo , Animais , Sequência de Bases , Linhagem Celular , DNA Nucleotidiltransferases/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Ácido Glutâmico/metabolismo , Camundongos Endogâmicos C57BL , Mutação/genética , Optogenética , Fenótipo
10.
Neuron ; 105(5): 909-920.e5, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-31879163

RESUMO

Dopamine neurons of the ventral tegmental area (VTA) regulate reward association and motivation. It remains unclear whether there are distinct dopamine populations to mediate these functions. Using mouse genetics, we isolated two populations of dopamine-producing VTA neurons with divergent projections to the nucleus accumbens (NAc) core and shell. Inhibition of VTA-core-projecting neurons disrupted Pavlovian reward learning, and activation of these cells promoted the acquisition of an instrumental response. VTA-shell-projecting neurons did not regulate Pavlovian reward learning and could not facilitate acquisition of an instrumental response, but their activation could drive robust responding in a previously learned instrumental task. Both populations are activated simultaneously by cues, actions, and rewards, and this co-activation is required for robust reinforcement of behavior. Thus, there are functionally distinct dopamine populations in the VTA for promoting motivation and reward association, which operate on the same timescale to optimize behavioral reinforcement.


Assuntos
Associação , Condicionamento Clássico/fisiologia , Neurônios Dopaminérgicos/fisiologia , Motivação , Núcleo Accumbens/fisiologia , Recompensa , Área Tegmentar Ventral/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Dependovirus , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Vetores Genéticos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Vias Neurais/fisiologia , Núcleo Accumbens/citologia , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Reforço Psicológico , Área Tegmentar Ventral/citologia
11.
Nat Med ; 24(4): 438-449, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29529016

RESUMO

Memories become less precise and generalized over time as memory traces reorganize in hippocampal-cortical networks. Increased time-dependent loss of memory precision is characterized by an overgeneralization of fear in individuals with post-traumatic stress disorder (PTSD) or age-related cognitive impairments. In the hippocampal dentate gyrus (DG), memories are thought to be encoded by so-called 'engram-bearing' dentate granule cells (eDGCs). Here we show, using rodents, that contextual fear conditioning increases connectivity between eDGCs and inhibitory interneurons (INs) in the downstream hippocampal CA3 region. We identify actin-binding LIM protein 3 (ABLIM3) as a mossy-fiber-terminal-localized cytoskeletal factor whose levels decrease after learning. Downregulation of ABLIM3 expression in DGCs was sufficient to increase connectivity with CA3 stratum lucidum INs (SLINs), promote parvalbumin (PV)-expressing SLIN activation, enhance feedforward inhibition onto CA3 and maintain a fear memory engram in the DG over time. Furthermore, downregulation of ABLIM3 expression in DGCs conferred conditioned context-specific reactivation of memory traces in hippocampal-cortical and amygdalar networks and decreased fear memory generalization at remote (i.e., distal) time points. Consistent with the observation of age-related hyperactivity of CA3, learning failed to increase DGC-SLIN connectivity in 17-month-old mice, whereas downregulation of ABLIM3 expression was sufficient to restore DGC-SLIN connectivity, increase PV+ SLIN activation and improve the precision of remote memories. These studies exemplify a connectivity-based strategy that targets a molecular brake of feedforward inhibition in DG-CA3 and may be harnessed to decrease time-dependent memory generalization in individuals with PTSD and improve memory precision in aging individuals.


Assuntos
Giro Denteado/citologia , Generalização da Resposta , Memória de Longo Prazo , Inibição Neural , Envelhecimento/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Região CA3 Hipocampal/fisiologia , Regulação para Baixo , Potenciais Pós-Sinápticos Excitadores , Medo , Feminino , Células HEK293 , Humanos , Interneurônios/fisiologia , Proteínas com Domínio LIM/metabolismo , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo
12.
Biol Psychiatry ; 84(6): 401-412, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29478701

RESUMO

BACKGROUND: Studies in humans and nonhuman primates have identified a region of the dentate nucleus of the cerebellum, or the lateral cerebellar nucleus (LCN) in rodents, activated during performance of cognitive tasks involving complex spatial and sequential planning. Whether such a subdivision exists in rodents is not known. Dopamine and its receptors, which are implicated in cognitive function, are present in the cerebellar nuclei, but their function is unknown. METHODS: Using viral and genetic strategies in mice, we examined cellular phenotypes of dopamine D1 receptor-positive (D1R+) cells in the LCN with whole-cell patch clamp recordings, messenger RNA profiling, and immunohistochemistry to examine D1R expression in mouse LCN and human dentate nucleus of the cerebellum. We used chemogenetics to inhibit D1R+ neurons and examined behaviors including spatial navigation, social recognition memory, prepulse inhibition of the acoustic startle reflex, response inhibition, and working memory to test the necessity of these neurons in these behaviors. RESULTS: We identified a population of D1R+ neurons that are localized to an anatomically distinct region of the LCN. We also observed D1R+ neurons in human dentate nucleus of the cerebellum, which suggests an evolutionarily conserved population of dopamine-receptive neurons in this region. The genetic, electrophysiological, and anatomical profile of mouse D1R neurons is consistent with a heterogeneous population of gamma-aminobutyric acidergic, and to a lesser extent glutamatergic, cell types. Selective inhibition of D1R+ LCN neurons impairs spatial navigation memory, response inhibition, working memory, and prepulse inhibition of the acoustic startle reflex. CONCLUSIONS: Collectively, these data demonstrate a functional link between genetically distinct neurons in the LCN and cognitive behaviors.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Núcleo Accumbens/fisiologia , Receptores de Dopamina D1/metabolismo , Comportamento Social , Animais , Comportamento Animal , Feminino , Masculino , Camundongos , Técnicas de Patch-Clamp , Reconhecimento Fisiológico de Modelo , Reflexo de Sobressalto , Memória Espacial
13.
Neuron ; 93(1): 164-178, 2017 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-28017470

RESUMO

Fear is a graded central motive state ranging from mild to intense. As threat intensity increases, fear transitions from discriminative to generalized. The circuit mechanisms that process threats of different intensity are not well resolved. Here, we isolate a unique population of locally projecting neurons in the central nucleus of the amygdala (CeA) that produce the neuropeptide corticotropin-releasing factor (CRF). CRF-producing neurons and CRF in the CeA are required for discriminative fear, but both are dispensable for generalized fear at high US intensities. Consistent with a role in discriminative fear, CRF neurons undergo plasticity following threat conditioning and selectively respond to threat-predictive cues. We further show that excitability of genetically isolated CRF-receptive (CRFR1) neurons in the CeA is potently enhanced by CRF and that CRFR1 signaling in the CeA is critical for discriminative fear. These findings demonstrate a novel CRF gain-control circuit and show separable pathways for graded fear processing.


Assuntos
Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/genética , Medo/fisiologia , Aprendizagem/fisiologia , Neurônios/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Tonsila do Cerebelo/fisiologia , Animais , Núcleo Central da Amígdala/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/fisiologia , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Transmissão Sináptica
14.
Cell Rep ; 16(2): 304-313, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27346361

RESUMO

Nearly all animals engage in a complex assortment of social behaviors that are essential for the survival of the species. In mammals, these behaviors are regulated by sub-nuclei within the hypothalamus, but the specific cell types within these nuclei responsible for coordinating behavior in distinct contexts are only beginning to be resolved. Here, we identify a population of neurons in the ventral premammillary nucleus of the hypothalamus (PMV) that are strongly activated in male intruder mice in response to a larger resident male but that are not responsive to females. Using a combination of molecular and genetic approaches, we demonstrate that these PMV neurons regulate intruder-specific male social behavior and social novelty recognition in a manner dependent on synaptic release of the excitatory neurotransmitter glutamate. These data provide direct evidence for a unique population of neurons that regulate social behaviors in specific contexts.


Assuntos
Comportamento Competitivo , Hipotálamo Posterior/citologia , Neurônios/fisiologia , Animais , Comportamento Animal , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Masculino , Camundongos
15.
J Neurosci ; 36(13): 3698-708, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27030756

RESUMO

Alcohol is the most commonly abused substance among adolescents, promoting the development of substance use disorders and compromised decision-making in adulthood. We have previously demonstrated, with a preclinical model in rodents, that adolescent alcohol use results in adult risk-taking behavior that positively correlates with phasic dopamine transmission in response to risky options, but the underlying mechanisms remain unknown. Here, we show that adolescent alcohol use may produce maladaptive decision-making through a disruption in dopamine network dynamics via increased GABAergic transmission within the ventral tegmental area (VTA). Indeed, we find that increased phasic dopamine signaling after adolescent alcohol use is attributable to a midbrain circuit, including the input from the pedunculopontine tegmentum to the VTA. Moreover, we demonstrate that VTA dopamine neurons from adult rats exhibit enhanced IPSCs after adolescent alcohol exposure corresponding to decreased basal dopamine levels in adulthood that negatively correlate with risk-taking. Building on these findings, we develop a model where increased inhibitory tone on dopamine neurons leads to a persistent decrease in tonic dopamine levels and results in a potentiation of stimulus-evoked phasic dopamine release that may drive risky choice behavior. Based on this model, we take a pharmacological approach to the reversal of risk-taking behavior through normalization of this pattern in dopamine transmission. These results isolate the underlying circuitry involved in alcohol-induced maladaptive decision-making and identify a novel therapeutic target. SIGNIFICANCE STATEMENT: One of the primary problems resulting from chronic alcohol use is persistent, maladaptive decision-making that is associated with ongoing addiction vulnerability and relapse. Indeed, studies with the Iowa Gambling Task, a standard measure of risk-based decision-making, have reliably shown that alcohol-dependent individuals make riskier, more maladaptive choices than nondependent individuals, even after periods of prolonged abstinence. Using a preclinical model, in the current work, we identify a selective disruption in dopamine network dynamics that may promote maladaptive decision-making after chronic adolescent alcohol use and demonstrate its pharmacological reversal in adulthood. Together, these results highlight a novel neural mechanism underlying heightened risk-taking behavior in alcohol-dependent individuals and provide a potential therapeutic target for further investigation.


Assuntos
Transtornos do Sistema Nervoso Induzidos por Álcool/complicações , Depressores do Sistema Nervoso Central/toxicidade , Tomada de Decisões/efeitos dos fármacos , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Etanol/toxicidade , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Estimulação Elétrica , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Microdiálise , Vias Neurais/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Assunção de Riscos , Área Tegmentar Ventral/patologia
16.
Nat Neurosci ; 19(5): 734-741, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27019015

RESUMO

In the face of starvation, animals will engage in high-risk behaviors that would normally be considered maladaptive. Starving rodents, for example, will forage in areas that are more susceptible to predators and will also modulate aggressive behavior within a territory of limited or depleted nutrients. The neural basis of these adaptive behaviors likely involves circuits that link innate feeding, aggression and fear. Hypothalamic agouti-related peptide (AgRP)-expressing neurons are critically important for driving feeding and project axons to brain regions implicated in aggression and fear. Using circuit-mapping techniques in mice, we define a disynaptic network originating from a subset of AgRP neurons that project to the medial nucleus of the amygdala and then to the principal bed nucleus of the stria terminalis, which suppresses territorial aggression and reduces contextual fear. We propose that AgRP neurons serve as a master switch capable of coordinating behavioral decisions relative to internal state and environmental cues.


Assuntos
Agressão/fisiologia , Proteína Relacionada com Agouti/fisiologia , Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Hipotálamo/fisiologia , Fragmentos de Peptídeos/fisiologia , Núcleos Septais/fisiologia , Inanição/fisiopatologia , Proteína Relacionada com Agouti/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Técnicas de Introdução de Genes , Hipotálamo/metabolismo , Masculino , Camundongos , Vias Neurais/metabolismo , Vias Neurais/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos/metabolismo , Núcleos Septais/metabolismo
17.
Elife ; 42015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26402461

RESUMO

The lateral amygdala (LA) acquires differential coding of predictive and non-predictive fear stimuli that is critical for proper fear memory assignment. The neurotransmitter dopamine is an important modulator of LA activity and facilitates fear memory formation, but whether dopamine neurons aid in the establishment of discriminative fear coding by the LA is unknown. NMDA-type glutamate receptors in dopamine neurons are critical for the prevention of generalized fear following an aversive experience, suggesting a potential link between a cell autonomous function of NMDAR in dopamine neurons and fear coding by the LA. Here, we utilized mice with a selective genetic inactivation functional NMDARs in dopamine neurons (DAT-NR1 KO mice) combined with behavior, in vivo electrophysiology, and ex vivo electrophysiology in LA neurons to demonstrate that plasticity underlying differential fear coding in the LA is regulated by NMDAR signaling in dopamine neurons and alterations in this plasticity is associated non-discriminative cued-fear responses.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Medo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Técnicas de Inativação de Genes , Masculino , Camundongos Knockout , Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato/genética
18.
Cell ; 162(2): 363-374, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26186190

RESUMO

Animals learn to avoid harmful situations by associating a neutral stimulus with a painful one, resulting in a stable threat memory. In mammals, this form of learning requires the amygdala. Although pain is the main driver of aversive learning, the mechanism that transmits pain signals to the amygdala is not well resolved. Here, we show that neurons expressing calcitonin gene-related peptide (CGRP) in the parabrachial nucleus are critical for relaying pain signals to the central nucleus of amygdala and that this pathway may transduce the affective motivational aspects of pain. Genetic silencing of CGRP neurons blocks pain responses and memory formation, whereas their optogenetic stimulation produces defensive responses and a threat memory. The pain-recipient neurons in the central amygdala expressing CGRP receptors are also critical for establishing a threat memory. The identification of the neural circuit conveying affective pain signals may be pertinent for treating pain conditions with psychiatric comorbidities.


Assuntos
Tonsila do Cerebelo/fisiologia , Vias Neurais , Neurônios/fisiologia , Dor/fisiopatologia , Animais , Comportamento Animal , Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Condicionamento Psicológico , Aprendizagem , Núcleos Parabraquiais/fisiologia , Precursores de Proteínas/genética
19.
Learn Mem ; 21(11): 575-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25320348

RESUMO

Dopamine is broadly implicated in fear-related processes, yet we know very little about signaling dynamics in these neurons during active fear conditioning. We describe the direct imaging of calcium signals of dopamine neurons during Pavlovian fear conditioning using fiber-optic confocal microscopy coupled with the genetically encoded calcium indicator GCaMP3. We observed calcium transients in a subset of dopamine neurons to an unconditioned fear stimulus on the first day of Pavlovian fear conditioning. On the second day, calcium transients occurred in response to conditioned and unconditioned stimuli. These results demonstrate plasticity in dopamine neuron calcium signals and the occurrence of activity-dependent processes in these neurons during fear conditioning.


Assuntos
Sinalização do Cálcio , Neurônios Dopaminérgicos/metabolismo , Medo/fisiologia , Plasticidade Neuronal , Área Tegmentar Ventral/metabolismo , Animais , Condicionamento Clássico/fisiologia , Eletrochoque , Camundongos
20.
Neuron ; 80(4): 997-1009, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24206670

RESUMO

The calcium-activated small conductance potassium channel SK3 plays an essential role in the regulation of dopamine neuron activity patterns. Here we demonstrate that expression of a human disease-related SK3 mutation (hSK3Δ) in dopamine neurons of mice disrupts the balance between tonic and phasic dopamine neuron activity. Expression of hSK3Δ suppressed endogenous SK currents, reducing coupling between SK channels and NMDA receptors (NMDARs) and increasing permissiveness for burst firing. Consistent with enhanced excitability of dopamine neurons, hSK3Δ increased evoked calcium signals in dopamine neurons in vivo and potentiated evoked dopamine release. Specific expression of hSK3Δ led to deficits in attention and sensory gating and heightened sensitivity to a psychomimetic drug. Sensory-motor alterations and psychomimetic sensitivity were recapitulated in a mouse model of transient, reversible dopamine neuron activation. These results demonstrate the cell-autonomous effects of a human ion channel mutation on dopamine neuron physiology and the impact of activity pattern disruption on behavior.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Atenção/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Dependovirus , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Alucinógenos/farmacologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , N-Metilaspartato/metabolismo , Desempenho Psicomotor/fisiologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/fisiologia , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia , Filtro Sensorial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA