Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 27(5): 2580-2589, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35418600

RESUMO

N-methyl-D-aspartate receptor (NMDAR) modulators have recently received increased attention as potential therapeutics for posttraumatic stress disorder (PTSD). Here, we tested a novel NMDAR-positive modulator, NYX-783, in the following two rodent models of PTSD: an auditory fear-conditioning model and a single-prolonged stress (SPS) model. We examined the ability of NYX-783 to reduce subsequent fear-based behaviors by measuring enhanced fear extinction and reduced spontaneous recovery (spontaneous return of fear) in male mice. NYX-783 administration significantly reduced spontaneous recovery in both PTSD models and enhanced fear extinction in the SPS model. Furthermore, NYX-783 increased the NMDA-induced inward currents of excitatory and inhibitory neurons in the infralimbic medial prefrontal cortex (IL mPFC) and that the GluN2B subunit of NMDARs on pyramidal neurons in the IL mPFC is required for its effect on spontaneous recovery. The downstream expression of brain-derived neurotrophic factor was required for NYX-783 to achieve its behavioral effect. These results elucidate the cellular targets of NYX-783 and the molecular mechanisms underlying the inhibition of spontaneous recovery. These preclinical findings support the hypothesis that NYX-783 may have therapeutic potential for PTSD treatment and may be particularly useful for inhibiting spontaneous recovery.


Assuntos
Medo , Receptores de N-Metil-D-Aspartato , Animais , Extinção Psicológica/fisiologia , Medo/fisiologia , Masculino , Camundongos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
2.
J Neurosci ; 40(24): 4727-4738, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32354856

RESUMO

Decades of research have shown that the NAc is a critical region influencing addiction, mood, and food consumption through its effects on reinforcement learning, motivation, and hedonic experience. Pharmacological studies have demonstrated that inhibition of the NAc shell induces voracious feeding, leading to the hypothesis that the inhibitory projections that emerge from the NAc normally act to restrict feeding. While much of this work has focused on projections to the lateral hypothalamus, the role of NAc projections to the VTA in the control food intake has been largely unexplored. Using a retrograde viral labeling technique and real-time monitoring of neural activity with fiber photometry, we find that medial NAc shell projections to the VTA (mNAc→VTA) are inhibited during food-seeking and food consumption in male mice. We also demonstrate that this circuit bidirectionally controls feeding: optogenetic activation of NAc projections to the VTA inhibits food-seeking and food intake (in both sexes), while optogenetic inhibition of this circuit potentiates food-seeking behavior. Additionally, we show that activity of the NAc to VTA pathway is necessary for adaptive inhibition of food intake in response to external cues. These data provide new insight into NAc control over feeding in mice, and contribute to an emerging literature elucidating the role of inhibitory midbrain feedback within the mesolimbic circuit.SIGNIFICANCE STATEMENT The medial NAc has long been known to control consummatory behavior, with particular focus on accumbens projections to the lateral hypothalamus. Conversely, NAc projections to the VTA have mainly been studied in the context of drug reward. We show that NAc projections to the VTA bidirectionally control food intake, consistent with a permissive role in feeding. Additionally, we show that this circuit is normally inactivated during consumption and food-seeking. Together, these findings elucidate how mesolimbic circuits control food consumption.


Assuntos
Comportamento Consumatório/fisiologia , Ingestão de Alimentos/fisiologia , Núcleo Accumbens/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Condicionamento Operante/fisiologia , Masculino , Camundongos , Atividade Motora/fisiologia , Vias Neurais/fisiologia , Optogenética , Recompensa
3.
J Neurosci ; 37(17): 4462-4471, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28336571

RESUMO

The ability to inhibit drinking is a significant challenge for recovering alcoholics, especially in the presence of alcohol-associated cues. Previous studies have demonstrated that the regulation of cue-guided alcohol seeking is mediated by the basolateral amygdala (BLA), nucleus accumbens (NAc), and medial prefrontal cortex (mPFC). However, given the high interconnectivity between these structures, it is unclear how mPFC projections to each subcortical structure, as well as projections between BLA and NAc, mediate alcohol-seeking behaviors. Here, we evaluate how cortico-striatal, cortico-amygdalar, and amygdalo-striatal projections control extinction and relapse in a rat model of alcohol seeking. Specifically, we used a combinatorial viral technique to express diphtheria toxin receptors in specific neuron populations based on their projection targets. We then used this strategy to create directionally selective ablations of three distinct pathways after acquisition of ethanol self-administration but before extinction and reinstatement. We demonstrate that ablation of mPFC neurons projecting to NAc, but not BLA, blocks cue-induced reinstatement of alcohol seeking and neither pathway is necessary for extinction of responding. Further, we show that ablating BLA neurons that project to NAc disrupts extinction of alcohol approach behaviors and attenuates reinstatement. Together, these data provide evidence that the mPFC→NAc pathway is necessary for cue-induced reinstatement of alcohol seeking, expand our understanding of how the BLA→NAc pathway regulates alcohol behavior, and introduce a new methodology for the manipulation of target-specific neural projections.SIGNIFICANCE STATEMENT The vast majority of recovering alcoholics will relapse at least once and understanding how the brain regulates relapse will be key to developing more effective behavior and pharmacological therapies for alcoholism. Given the high interconnectivity of cortical, striatal, and limbic structures that regulate alcohol intake, it has been difficult to disentangle how separate projections between them may control different aspects of these complex behaviors. Here, we demonstrate a new approach for noninvasively ablating each of these pathways and testing their necessity for both extinction and relapse. We show that inputs to the nucleus accumbens from medial prefrontal cortex and amygdala regulate alcohol-seeking behaviors differentially, adding to our understanding of the neural control of alcoholism.


Assuntos
Alcoolismo/fisiopatologia , Alcoolismo/psicologia , Tonsila do Cerebelo/fisiopatologia , Sinais (Psicologia) , Extinção Psicológica , Núcleo Accumbens/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Animais , Comportamento Animal , Condicionamento Operante/efeitos dos fármacos , Etanol/farmacologia , Masculino , Vias Neurais/fisiopatologia , Ratos , Ratos Sprague-Dawley , Recidiva
4.
Proc Natl Acad Sci U S A ; 112(19): 6188-93, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918363

RESUMO

Current treatments for major depressive disorder (MDD) have a time lag and are ineffective for a large number of patients. Development of novel pharmacological therapies requires a comprehensive understanding of the molecular events that contribute to MDD pathophysiology. Recent evidence points toward aberrant activity of synaptic proteins as a critical contributing factor. In the present studies, we used viral-mediated gene transfer to target a key mediator of activity-dependent synaptic protein synthesis downstream of mechanistic target of rapamycin complex 1 (mTORC1) known as p70 S6 kinase 1 (S6K1). Targeted delivery of two mutants of S6K1, constitutively active or dominant-negative, to the medial prefrontal cortex (mPFC) of rats allowed control of the mTORC1/S6K1 translational pathway. Our results demonstrate that increased expression of S6K1 in the mPFC produces antidepressant effects in the forced swim test without altering locomotor activity. Moreover, expression of active S6K1 in the mPFC blocked the anhedonia caused by chronic stress, resulting in a state of stress resilience. This antidepressant response was associated with increased neuronal complexity caused by enhanced S6K1 activity. Conversely, expression of dominant-negative S6K1 in the mPFC resulted in prodepressive behavior in the forced swim test and was sufficient to cause anhedonia in the absence of chronic stress exposure. Together, these data demonstrate a critical role for S6K1 activity in depressive behaviors, and suggest that pathways downstream of mTORC1 may underlie the pathophysiology and treatment of MDD.


Assuntos
Transtorno Depressivo Maior/metabolismo , Regulação Enzimológica da Expressão Gênica , Córtex Pré-Frontal/metabolismo , Proteínas Quinases S6 Ribossômicas/fisiologia , Animais , Antidepressivos/uso terapêutico , Comportamento Animal , Modelos Animais de Doenças , Ketamina/química , Masculino , Aprendizagem em Labirinto , Neurônios/metabolismo , Fenótipo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/genética , Transdução de Sinais , Sirolimo/química , Natação
5.
Proc Natl Acad Sci U S A ; 112(3): 893-8, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25561540

RESUMO

Gilles de la Tourette syndrome (TS) is characterized by tics, which are transiently worsened by stress, acute administration of dopaminergic drugs, and by subtle deficits in motor coordination and sensorimotor gating. It represents the most severe end of a spectrum of tic disorders that, in aggregate, affect ∼ 5% of the population. Available treatments are frequently inadequate, and the pathophysiology is poorly understood. Postmortem studies have revealed a reduction in specific striatal interneurons, including the large cholinergic interneurons, in severe disease. We tested the hypothesis that this deficit is sufficient to produce aspects of the phenomenology of TS, using a strategy for targeted, specific cell ablation in mice. We achieved ∼ 50% ablation of the cholinergic interneurons of the striatum, recapitulating the deficit observed in patients postmortem, without any effect on GABAergic markers or on parvalbumin-expressing fast-spiking interneurons. Interneuron ablation in the dorsolateral striatum (DLS), corresponding roughly to the human putamen, led to tic-like stereotypies after either acute stress or d-amphetamine challenge; ablation in the dorsomedial striatum, in contrast, did not. DLS interneuron ablation also led to a deficit in coordination on the rotorod, but not to any abnormalities in prepulse inhibition, a measure of sensorimotor gating. These results support the causal sufficiency of cholinergic interneuron deficits in the DLS to produce some, but not all, of the characteristic symptoms of TS.


Assuntos
Corpo Estriado/patologia , Toxina Diftérica/farmacologia , Interneurônios/citologia , Receptores Colinérgicos/metabolismo , Síndrome de Tourette/patologia , Potenciais de Ação , Animais , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Síndrome de Tourette/psicologia
6.
Proc Natl Acad Sci U S A ; 112(26): 8106-11, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26056286

RESUMO

Ketamine produces rapid and sustained antidepressant actions in depressed patients, but the precise cellular mechanisms underlying these effects have not been identified. Here we determined if modulation of neuronal activity in the infralimbic prefrontal cortex (IL-PFC) underlies the antidepressant and anxiolytic actions of ketamine. We found that neuronal inactivation of the IL-PFC completely blocked the antidepressant and anxiolytic effects of systemic ketamine in rodent models and that ketamine microinfusion into IL-PFC reproduced these behavioral actions of systemic ketamine. We also found that optogenetic stimulation of the IL-PFC produced rapid and long-lasting antidepressant and anxiolytic effects and that these effects are associated with increased number and function of spine synapses of layer V pyramidal neurons. The results demonstrate that ketamine infusions or optogenetic stimulation of IL-PFC are sufficient to produce long-lasting antidepressant behavioral and synaptic responses similar to the effects of systemic ketamine administration.


Assuntos
Antidepressivos/farmacologia , Ketamina/farmacologia , Sistema Límbico/efeitos dos fármacos , Optogenética , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Sistema Límbico/fisiopatologia , Masculino , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Sprague-Dawley
7.
Proc Natl Acad Sci U S A ; 111(32): 11876-81, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25071172

RESUMO

Prolyl endopeptidase (PREP) has been implicated in neuronal functions. Here we report that hypothalamic PREP is predominantly expressed in the ventromedial nucleus (VMH), where it regulates glucose-induced neuronal activation. PREP knockdown mice (Prep(gt/gt)) exhibited glucose intolerance, decreased fasting insulin, increased fasting glucagon levels, and reduced glucose-induced insulin secretion compared with wild-type controls. Consistent with this, central infusion of a specific PREP inhibitor, S17092, impaired glucose tolerance and decreased insulin levels in wild-type mice. Arguing further for a central mode of action of PREP, isolated pancreatic islets showed no difference in glucose-induced insulin release between Prep(gt/gt) and wild-type mice. Furthermore, hyperinsulinemic euglycemic clamp studies showed no difference between Prep(gt/gt) and wild-type control mice. Central PREP regulation of insulin and glucagon secretion appears to be mediated by the autonomic nervous system because Prep(gt/gt) mice have elevated sympathetic outflow and norepinephrine levels in the pancreas, and propranolol treatment reversed glucose intolerance in these mice. Finally, re-expression of PREP by bilateral VMH injection of adeno-associated virus-PREP reversed the glucose-intolerant phenotype of the Prep(gt/gt) mice. Taken together, our results unmask a previously unknown player in central regulation of glucose metabolism and pancreatic function.


Assuntos
Glucagon/metabolismo , Hipotálamo/enzimologia , Insulina/metabolismo , Serina Endopeptidases/metabolismo , Animais , Glicemia/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Técnica Clamp de Glucose , Intolerância à Glucose/enzimologia , Intolerância à Glucose/etiologia , Hipotálamo/fisiologia , Indóis/farmacologia , Secreção de Insulina , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/genética , Pâncreas/metabolismo , Fosforilação , Prolil Oligopeptidases , Receptor de Insulina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Inibidores de Serina Proteinase/farmacologia , Tiazolidinas/farmacologia , Proteína Desacopladora 1 , Núcleo Hipotalâmico Ventromedial/enzimologia , Núcleo Hipotalâmico Ventromedial/fisiologia
8.
Proc Natl Acad Sci U S A ; 109(50): 20726-31, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23185016

RESUMO

Temporal control, or how organisms guide movements in time to achieve behavioral goals, depends on dopamine signaling. The medial prefrontal cortex controls many goal-directed behaviors and receives dopaminergic input primarily from the midbrain ventral tegmental area. However, this system has never been linked with temporal control. Here, we test the hypothesis that dopaminergic projections from the ventral tegmental area to the prefrontal cortex influence temporal control. Rodents were trained to perform a fixed-interval timing task with an interval of 20 s. We report several results: first, that decreasing dopaminergic neurotransmission using virally mediated RNA interference of tyrosine hydroxylase impaired temporal control, and second that pharmacological disruption of prefrontal D1 dopamine receptors, but not D2 dopamine receptors, impaired temporal control. We then used optogenetics to specifically and selectively manipulate prefrontal neurons expressing D1 dopamine receptors during fixed-interval timing performance. Selective inhibition of D1-expressing prefrontal neurons impaired fixed-interval timing, whereas stimulation made animals more efficient during task performance. These data provide evidence that ventral tegmental dopaminergic projections to the prefrontal cortex influence temporal control via D1 receptors. The results identify a critical circuit for temporal control of behavior that could serve as a target for the treatment of dopaminergic diseases.


Assuntos
Córtex Pré-Frontal/fisiologia , Receptores de Dopamina D1/fisiologia , Animais , Sequência de Bases , Comportamento Animal/fisiologia , Relógios Biológicos/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Vias Neurais/fisiologia , Optogenética , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Ratos , Recompensa , Transdução de Sinais , Transmissão Sináptica , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/fisiologia , Área Tegmentar Ventral/fisiologia
9.
Proc Natl Acad Sci U S A ; 109(50): 20714-9, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23185000

RESUMO

Stressor exposure biases decision-making strategies from those based on the relationship between actions and their consequences to others restricted by stimulus-response associations. Chronic stressor exposure also desensitizes glucocorticoid receptors (GR) and diminishes motivation to acquire food reinforcement, although causal relationships are largely not established. We show that a history of chronic exposure to the GR ligand corticosterone or acute posttraining GR blockade with RU38486 makes rodents less able to perform actions based on their consequences. Thus, optimal GR binding is necessary for the consolidation of new response-outcome learning. In contrast, medial prefrontal (but not striatal) BDNF can account for stress-related amotivation, in that selective medial prefrontal cortical Bdnf knockdown decreases break-point ratios in a progressive-ratio task. Knockdown also increases vulnerability to RU38486. Despite the role of BDNF in dendritic spine reorganization, deep-layer spine remodeling does not obviously parallel progressive-ratio response patterns, but treatment with the Na(+)-channel inhibitor riluzole reverses corticosteroid-induced motivational deficits and restores prefrontal BDNF expression after corticosterone. We argue that when prefrontal neurotrophin systems are compromised, and GR-mediated hypothalamic-pituitary-adrenal axis feedback is desensitized (as in the case of chronic stress hormone exposure), amotivation and inflexible maladaptive response strategies that contribute to stress-related mood disorders result.


Assuntos
Comportamento Animal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores de Glucocorticoides/fisiologia , Afeto/efeitos dos fármacos , Afeto/fisiologia , Amitriptilina/farmacologia , Animais , Antidepressivos Tricíclicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/genética , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Corticosterona/farmacologia , Corticosterona/fisiologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mifepristona/farmacologia , Motivação/efeitos dos fármacos , Motivação/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inibidores , Estresse Fisiológico
10.
Transl Psychiatry ; 14(1): 90, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38346984

RESUMO

Recent studies have implicated the endogenous opioid system in the antidepressant actions of ketamine, but the underlying mechanisms remain unclear. We used a combination of pharmacological, behavioral, and molecular approaches in rats to test the contribution of the prefrontal endogenous opioid system to the antidepressant-like effects of a single dose of ketamine. Both the behavioral actions of ketamine and their molecular correlates in the medial prefrontal cortex (mPFC) are blocked by acute systemic administration of naltrexone, a competitive opioid receptor antagonist. Naltrexone delivered directly into the mPFC similarly disrupts the behavioral effects of ketamine. Ketamine treatment rapidly increases levels of ß-endorphin and the expression of the µ-opioid receptor gene (Oprm1) in the mPFC, and the expression of gene that encodes proopiomelanocortin, the precursor of ß-endorphin, in the hypothalamus, in vivo. Finally, neutralization of ß-endorphin in the mPFC using a specific antibody prior to ketamine treatment abolishes both behavioral and molecular effects. Together, these findings indicate that presence of ß-endorphin and activation of opioid receptors in the mPFC are required for the antidepressant-like actions of ketamine.


Assuntos
Ketamina , Ratos , Animais , Analgésicos Opioides/farmacologia , beta-Endorfina/metabolismo , beta-Endorfina/farmacologia , Naltrexona/farmacologia , Naltrexona/metabolismo , Antidepressivos , Córtex Pré-Frontal/metabolismo
11.
bioRxiv ; 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38645212

RESUMO

Problematic opioid use that emerges in a subset of individuals may be due to pre-existing disruptions in the biobehavioral mechanisms that regulate drug use. The identity of these mechanisms is not known, but emerging evidence suggests that suboptimal decision-making that is observable prior to drug use may contribute to the pathology of addiction and, notably, serve as a powerful phenotype for interrogating biologically based differences in opiate-taking behaviors. The current study investigated the relationship between decision-making phenotypes and opioid-taking behaviors in male and female Long Evans rats. Adaptive decision-making processes were assessed using a probabilistic reversal-learning task and oxycodone- (or vehicle, as a control) taking behaviors assessed for 32 days using a saccharin fading procedure that promoted dynamic intake of oxycodone. Tests of motivation, extinction, and reinstatement were also performed. Computational analyses of decision-making and opioid-taking behaviors revealed that attenuated reward-guided decision-making was associated with greater self-administration of oxycodone and addiction-relevant behaviors. Moreover, pre-existing impairments in reward-guided decision-making observed in female rats was associated with greater oxycodone use and addiction-relevant behaviors when compared to males. These results provide new insights into the biobehavioral mechanisms that regulate opiate-taking behaviors and offer a novel phenotypic approach for interrogating sex differences in addiction susceptibility and opioid use disorders.

12.
Eur J Neurosci ; 38(3): 2382-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23651226

RESUMO

The orbitofrontal cortex (oPFC) sends substantial projections to the ventrolateral striatum and aspects of the nucleus accumbens that are, functionally, poorly understood. This is despite probable cortico-striatal involvement in multiple diseases such as addiction and obsessive-compulsive disorder. Here we surgically disconnected the oPFC from the ventrolateral striatum using unilateral asymmetric lesions in mice and classified instrumental decision-making strategies. Mice with symmetric lesions that spared one oPFC-striatal network served as controls. As a complementary approach, we selectively knocked down Brain-derived neurotrophic factor (Bdnf) bilaterally in the oPFC and ascertained behavioral and neurobiological consequences within the downstream striatum. oPFC-striatal disconnection and oPFC Bdnf knockdown blocked sensitivity to outcome-predictive relationships in both food-reinforced and cocaine-associated settings. Bdnf knockdown simultaneously regulated striatal BDNF expression, and striatal c-Fos predicted sensitivity to action-outcome associative contingencies. Previous evidence strongly implicates the dorsolateral striatum in stimulus-response habit formation. Our findings thus provide novel evidence for functional compartmentalisation within the lateral striatum, with the dorsal compartment subserving classical stimulus-response habit systems and a ventral compartment coordinating outcome-based decision-making via oPFC interactions. This compartmentalisation may apply to both 'natural', as in the case of food-reinforced behavior, and 'pathological', as in the case of cocaine-seeking, contexts.


Assuntos
Corpo Estriado/fisiologia , Tomada de Decisões/fisiologia , Lobo Frontal/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Condicionamento Operante/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/fisiologia
13.
Neuropsychopharmacology ; 48(3): 489-497, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36100654

RESUMO

Clinical investigations suggest involvement of the metabotropic glutamate receptor 5 (mGluR5) in the pathophysiology of fear learning that underlies trauma-related disorders. Here, we utilized a 4-day fear learning paradigm combined with positron emission tomography (PET) to examine the relationship between mGluR5 availability and differences in the response of rats to repeated footshock exposure (FE). Specifically, on day 1, male (n = 16) and female (n = 12) rats received 15 footshocks and were compared with control rats who did not receive footshocks (n = 7 male; n = 4 female). FE rats were classified as low responders (LR) or high responders (HR) based on freezing to the context the following day (day 2). PET with [18F]FPEB was used to calculate regional mGluR5 binding potential (BPND) at two timepoints: prior to FE (i.e., baseline), and post-behavioral testing. Additionally, we used an unbiased proteomics approach to assess group and sex differences in prefrontal cortex (PFC) protein expression. Post-behavioral testing we observed decreased BPND in LR females, but increased BPND in HR males relative to baseline. Further, individuals displaying the greatest freezing during the FE context memory test had the largest increases in PFC BPND. Males and females displayed unique post-test molecular profiles: in males, the greatest differences were between FE and CON, including upregulation of mGluR5 and related molecular networks in FE, whereas the greatest differences among females were between the LR and HR groups. These findings suggest greater mGluR5 availability increases following footshock exposure may be related to greater contextual fear memory. Results additionally reveal sex differences in the molecular response to footshock, including differential involvement of mGluR5-related molecular networks.


Assuntos
Receptor de Glutamato Metabotrópico 5 , Animais , Feminino , Masculino , Ratos , Tomografia por Emissão de Pósitrons/métodos , Receptor de Glutamato Metabotrópico 5/metabolismo , Fatores Sexuais
14.
Nat Metab ; 5(6): 1059-1072, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37308722

RESUMO

Post-ingestive nutrient signals to the brain regulate eating behaviour in rodents, and impaired responses to these signals have been associated with pathological feeding behaviour and obesity. To study this in humans, we performed a single-blinded, randomized, controlled, crossover study in 30 humans with a healthy body weight (females N = 12, males N = 18) and 30 humans with obesity (females N = 18, males N = 12). We assessed the effect of intragastric glucose, lipid and water (noncaloric isovolumetric control) infusions on the primary endpoints cerebral neuronal activity and striatal dopamine release, as well as on the secondary endpoints plasma hormones and glucose, hunger scores and caloric intake. To study whether impaired responses in participants with obesity would be partially reversible with diet-induced weight loss, imaging was repeated after 10% diet-induced weight loss. We show that intragastric glucose and lipid infusions induce orosensory-independent and preference-independent, nutrient-specific cerebral neuronal activity and striatal dopamine release in lean participants. In contrast, participants with obesity have severely impaired brain responses to post-ingestive nutrients. Importantly, the impaired neuronal responses are not restored after diet-induced weight loss. Impaired neuronal responses to nutritional signals may contribute to overeating and obesity, and ongoing resistance to post-ingestive nutrient signals after significant weight loss may in part explain the high rate of weight regain after successful weight loss.


Assuntos
Dopamina , Obesidade , Masculino , Feminino , Humanos , Estudos Cross-Over , Redução de Peso , Encéfalo , Nutrientes , Glucose , Lipídeos
15.
Am J Physiol Endocrinol Metab ; 303(4): E496-503, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22693203

RESUMO

Leptin receptor (LepRb) signaling in the hindbrain is required for energy balance control. Yet the specific hindbrain neurons and the behavioral processes mediating energy balance control by hindbrain leptin signaling are unknown. Studies here employ genetic [adeno-associated virally mediated RNA interference (AAV-RNAi)] and pharmacological methodologies to specify the neurons and the mechanisms through which hindbrain LepRb signaling contributes to the control of food intake. Results show that AAV-RNAi-mediated LepRb knockdown targeting a region encompassing the mNTS and area postrema (AP) (mNTS/AP LepRbKD) increases overall cumulative food intake by increasing the size of spontaneous meals. Other results show that pharmacological hindbrain leptin delivery and RNAi-mediated mNTS/AP LepRb knockdown increased and decreased the intake-suppressive effects of intraduodenal nutrient infusion, respectively. These meal size and intestinally derived signal amplification effects are likely mediated by LepRb signaling in the mNTS and not the AP, since 4th icv and mNTS parenchymal leptin (0.5 µg) administration reduced food intake, whereas this dose did not influence food intake when injected into the AP. Overall, these findings deepen the understanding of the distributed neuronal systems and behavioral mechanisms that mediate the effects of leptin receptor signaling on the control of food intake.


Assuntos
Ingestão de Alimentos/fisiologia , Intestinos/fisiologia , Receptores para Leptina/fisiologia , Saciação/fisiologia , Transdução de Sinais/fisiologia , Núcleo Solitário/fisiologia , Animais , Área Postrema/efeitos dos fármacos , Área Postrema/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Leptina/farmacologia , Masculino , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/genética , Saciação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos
16.
Biol Psychiatry ; 91(9): 841-852, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35279280

RESUMO

BACKGROUND: Stress produces differential behavioral responses through select molecular modifications to specific neurocircuitry elements. The orexin (Orx) system targets key components of this neurocircuitry in the basolateral amygdala (BLA). METHODS: We assessed the contribution of intra-BLA Orx1 receptors (Orx1Rs) in the expression of stress-induced phenotypes of mice. Using the Stress Alternatives Model, a social stress paradigm that produces two behavioral phenotypes, we characterized the role of intra-BLA Orx1R using acute pharmacological inhibition (SB-674042) and genetic knockdown (AAV-U6-Orx1R-shRNA) strategies. RESULTS: In the BLA, we observed that Orx1R (Hcrtr1) messenger RNA is predominantly expressed in CamKIIα+ glutamatergic neurons and rarely in GABAergic (gamma-aminobutyric acidergic) cells. While there is a slight overlap in Hcrtr1 and Orx2 receptor (Hcrtr2) messenger RNA expression in the BLA, we find that these receptors are most often expressed in separate cells. Antagonism of intra-BLA Orx1R after phenotype formation shifted behavioral expression from stress-sensitive (Stay) to stress-resilient (Escape) responses, an effect that was mimicked by genetic knockdown. Acute inhibition of Orx1R in the BLA also reduced contextual and cued fear freezing responses in Stay animals. This phenotype-specific behavioral change was accompanied by biased molecular transcription favoring Hcrtr2 over Hcrtr1 and Mapk3 over Plcb1 cell signaling cascades and enhanced Bdnf messenger RNA. CONCLUSIONS: Functional reorganization of intra-BLA gene expression is produced by antagonism of Orx1R, which promotes elevated Hcrtr2, greater Mapk3, and increased Bdnf expression. Together, these results provide evidence for a receptor-driven mechanism that balances pro- and antistress responses within the BLA.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Receptores de Orexina , Animais , Ansiedade/metabolismo , Complexo Nuclear Basolateral da Amígdala/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Camundongos , Receptores de Orexina/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
17.
J Neurosci ; 30(24): 8263-73, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20554878

RESUMO

The lateral hypothalamus and the nucleus accumbens shell (AcbSh) are brain regions important for food intake. The AcbSh contains high levels of receptor for melanin-concentrating hormone (MCH), a lateral hypothalamic peptide critical for feeding and metabolism. MCH receptor (MCHR1) activation in the AcbSh increases food intake, while AcbSh MCHR1 blockade reduces feeding. Here biochemical and cellular mechanisms of MCH action in the rodent AcbSh are described. A reduction of phosphorylation of GluR1 at serine 845 (pSer(845)) is shown to occur after both pharmacological and genetic manipulations of MCHR1 activity. These changes depend upon signaling through G(i/o), and result in decreased surface expression of GluR1-containing AMPA receptors (AMPARs). Electrophysiological analysis of medium spiny neurons (MSNs) in the AcbSh revealed decreased amplitude of AMPAR-mediated synaptic events (mEPSCs) with MCH treatment. In addition, MCH suppressed action potential firing MSNs through K(+) channel activation. Finally, in vivo recordings confirmed that MCH reduces neuronal cell firing in the AcbSh in freely moving animals. The ability of MCH to reduce cell firing in the AcbSh is consistent with a general model from other pharmacological and electrophysiological studies whereby reduced AcbSh neuronal firing leads to food intake. The current work integrates the hypothalamus into this model, providing biochemical and cellular mechanisms whereby metabolic and limbic signals converge to regulate food intake.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Núcleo Accumbens/fisiologia , Hormônios Hipofisários/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Compostos de Bário/farmacologia , Biotina/análogos & derivados , Biotina/metabolismo , Cloretos/farmacologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/farmacologia , Hipotálamo/citologia , Técnicas In Vitro , Masculino , Melaninas/genética , Melaninas/farmacologia , Camundongos , Camundongos Transgênicos , Vias Neurais/fisiologia , Neurônios/classificação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Núcleo Accumbens/citologia , Técnicas de Patch-Clamp/métodos , Hormônios Hipofisários/genética , Hormônios Hipofisários/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Long-Evans , Ratos Wistar , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Serina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
18.
Front Neuroendocrinol ; 31(1): 104-12, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19836414

RESUMO

Recent evidence has emerged demonstrating that metabolic hormones such as ghrelin and leptin can act on ventral tegmental area (VTA) midbrain dopamine neurons to influence feeding. The VTA is the origin of mesolimbic dopamine neurons that project to the nucleus accumbens (NAc) to influence behavior. While blockade of dopamine via systemic antagonists or targeted gene delete can impair food intake, local NAc dopamine manipulations have little effect on food intake. Notably, non-dopaminergic manipulations in the VTA and NAc produce more consistent effects on feeding and food choice. More recent genetic evidence supports a role for the substantia nigra-striatal dopamine pathways in food intake, while the VTA-NAc circuit is more likely involved in higher-order aspects of food acquisition, such as motivation and cue associations. This rich and complex literature should be considered in models of how peripheral hormones influence feeding behavior via action on the midbrain circuits.


Assuntos
Dopamina/fisiologia , Ingestão de Alimentos/fisiologia , Hormônios Peptídicos/fisiologia , Animais , Corpo Estriado/fisiologia , Grelina/fisiologia , Humanos , Leptina/fisiologia , Neurônios/fisiologia , Núcleo Accumbens/fisiologia , Transdução de Sinais , Substância Negra/fisiologia , Área Tegmentar Ventral/fisiologia
19.
Nat Med ; 9(12): 1539-44, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14634645

RESUMO

Conditional mutant techniques that allow spatial and temporal control over gene expression can be used to create mice with restricted genetic modifications. These mice serve as powerful disease models in which gene function in adult tissues can be specifically dissected. Current strategies for conditional genetic manipulation are inefficient, however, and often lack sufficient spatial control. Here we use viral-mediated RNA interference (RNAi) to generate a specific knockdown of Th, the gene encoding the dopamine synthesis enzyme tyrosine hydroxylase, within midbrain neurons of adult mice. This localized gene knockdown resulted in behavioral changes, including a motor performance deficit and reduced response to a psychostimulant. These results underscore the potential of using viral-mediated RNAi for the rapid production and testing of new genetic disease models. Similar strategies may be used in other model species, and may ultimately find applications in human gene therapy.


Assuntos
Encéfalo/metabolismo , Marcação de Genes/métodos , Interferência de RNA , Animais , Sequência de Bases , Comportamento Animal , DNA/genética , Dependovirus/genética , Expressão Gênica , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tirosina 3-Mono-Oxigenase/biossíntese , Tirosina 3-Mono-Oxigenase/genética
20.
Nat Neurosci ; 10(8): 1029-37, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17618281

RESUMO

A single exposure to cocaine rapidly induces the brief activation of several immediate early genes, but the role of such short-term regulation in the enduring consequences of cocaine use is poorly understood. We found that 4 h of intravenous cocaine self-administration in rats induced a transient increase in brain-derived neurotrophic factor (BDNF) and activation of TrkB-mediated signaling in the nucleus accumbens (NAc). Augmenting this dynamic regulation with five daily NAc BDNF infusions caused enduring increases in cocaine self-administration, and facilitated relapse to cocaine seeking in withdrawal. In contrast, neutralizing endogenous BDNF regulation with intra-NAc infusions of antibody to BDNF subsequently reduced cocaine self-administration and attenuated relapse. Using localized inducible BDNF knockout in mice, we found that BDNF originating from NAc neurons was necessary for maintaining increased cocaine self-administration. These findings suggest that dynamic induction and release of BDNF from NAc neurons during cocaine use promotes the development and persistence of addictive behavior.


Assuntos
Anestésicos Locais/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cocaína/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Reforço Psicológico , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Accumbens/citologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Autoadministração/métodos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA