Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Addict Biol ; 26(3): e12961, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32820590

RESUMO

Individuals suffering from substance use disorder often experience relapse events that are attributed to drug craving. Insular cortex (IC) function is implicated in processing drug-predictive cues and is thought to be a critical substrate for drug craving, but the downstream neural circuit effectors of the IC that mediate reward processing are poorly described. Here, we uncover the functional connectivity of an IC projection to the ventral bed nucleus of the stria terminalis (vBNST), a portion of the extended amygdala that has been previously shown to modulate dopaminergic activity within the ventral tegmental area (VTA), and investigate the role of this pathway in reward-related behaviors. We utilized ex vivo slice electrophysiology and in vivo optogenetics to examine the functional connectivity of the IC-vBNST projection and bidirectionally control IC-vBNST terminals in various reward-related behavioral paradigms. We hypothesized that the IC recruits mesolimbic dopamine signaling by activating VTA-projecting, vBNST neurons. Using slice electrophysiology, we found that the IC sends a glutamatergic projection onto vBNST-VTA neurons. Photoactivation of IC-vBNST terminals was sufficient to reinforce behavior in a dopamine-dependent manner. Moreover, silencing the IC-vBNST projection was aversive and resulted in anxiety-like behavior without affecting food consumption. This work provides a potential mechanism by which the IC processes exteroceptive triggers that are predictive of reward.


Assuntos
Comportamento Animal/fisiologia , Córtex Cerebral/fisiologia , Dopamina/metabolismo , Núcleos Septais/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Ansiedade/fisiopatologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Recompensa , Área Tegmentar Ventral/fisiologia
2.
Nature ; 496(7444): 224-8, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23515155

RESUMO

The co-morbidity of anxiety and dysfunctional reward processing in illnesses such as addiction and depression suggests that common neural circuitry contributes to these disparate neuropsychiatric symptoms. The extended amygdala, including the bed nucleus of the stria terminalis (BNST), modulates fear and anxiety, but also projects to the ventral tegmental area (VTA), a region implicated in reward and aversion, thus providing a candidate neural substrate for integrating diverse emotional states. However, the precise functional connectivity between distinct BNST projection neurons and their postsynaptic targets in the VTA, as well as the role of this circuit in controlling motivational states, have not been described. Here we record and manipulate the activity of genetically and neurochemically identified VTA-projecting BNST neurons in freely behaving mice. Collectively, aversive stimuli exposure produced heterogeneous firing patterns in VTA-projecting BNST neurons. By contrast, in vivo optically identified glutamatergic projection neurons displayed a net enhancement of activity to aversive stimuli, whereas the firing rate of identified GABAergic (γ-aminobutyric acid-containing) projection neurons was suppressed. Channelrhodopsin-2-assisted circuit mapping revealed that both BNST glutamatergic and GABAergic projections preferentially innervate postsynaptic non-dopaminergic VTA neurons, thus providing a mechanistic framework for in vivo circuit perturbations. In vivo photostimulation of BNST glutamatergic projections resulted in aversive and anxiogenic behavioural phenotypes. Conversely, activation of BNST GABAergic projections produced rewarding and anxiolytic phenotypes, which were also recapitulated by direct inhibition of VTA GABAergic neurons. These data demonstrate that functionally opposing BNST to VTA circuits regulate rewarding and aversive motivational states, and may serve as a crucial circuit node for bidirectionally normalizing maladaptive behaviours.


Assuntos
Tonsila do Cerebelo/fisiologia , Motivação/fisiologia , Animais , Ansiedade/fisiopatologia , Aprendizagem da Esquiva , Comportamento Animal/fisiologia , Channelrhodopsins , Sinais (Psicologia) , Eletrochoque , Neurônios GABAérgicos/metabolismo , Glutamina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética , Fenótipo , Recompensa , Núcleos Septais/fisiologia , Área Tegmentar Ventral/fisiologia
3.
J Neurosci ; 36(42): 10831-10842, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798138

RESUMO

The neural circuitry underlying mammalian reward behaviors involves several distinct nuclei throughout the brain. It is widely accepted that the midbrain dopamine (DA) neurons are critical for the reward-related behaviors. Recent studies have shown that the centromedial nucleus of the amygdala (CeMA) has a distinct role in regulating reward-related behaviors. However, the CeMA and ventromedial PFC (vmPFC) interaction in reward regulation remains poorly understood. Here, we identify and dissect a GABAergic projection that originates in the CeMA and terminates in the vmPFC (VGat-CreCeMA-vmPFC) using viral-vector-mediated, cell-type-specific optogenetic techniques in mice. Pathway-specific optogenetic activation of the VGat-CreCeMA-vmPFC circuit in awake, behaving animals produced a positive, reward-like phenotype in real-time place preference and increased locomotor activity in open-field testing. In sucrose operant conditioning, the photoactivation of these terminals increased nose-poking effort with no effect on licking behavior and robustly facilitated the extinction of operant behavior. However, photoactivation of these terminals did not induce self-stimulation in the absence of an external reward. The results described here suggest that the VGat-CreCeMA-vmPFC projection acts to modulate existing reward-related behaviors. SIGNIFICANCE STATEMENT: Many studies have shown that the interactions between the centromedial nucleus of the amygdala (CeMA) and ventromedial PFC (vmPFC) have critical roles for emotional regulation. However, most studies have associated this circuit with fear and anxiety behaviors and emphasized top-down processing from vmPFC to CeMA. Here, we provide new evidence for bottom-up CeMA to vmPFC influence on reward-related behaviors. Although previous work implicated the CeMA in incentive salience, our results isolate the investigation to a specific CeMA GABAergic projection to the vmPFC. This long-range GABAergic interaction between amygdala and frontal cortex adds a new dimension to the complex regulation of reward-related behaviors.


Assuntos
Tonsila do Cerebelo/fisiologia , Comportamento Animal/fisiologia , Córtex Pré-Frontal/fisiologia , Recompensa , Ácido gama-Aminobutírico/fisiologia , Animais , Condicionamento Operante/efeitos dos fármacos , Extinção Psicológica , Masculino , Camundongos , Atividade Motora , Vias Neurais/fisiologia , Optogenética , Autoestimulação , Sacarose/farmacologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
4.
Nature ; 475(7356): 377-80, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21716290

RESUMO

The basolateral amygdala (BLA) has a crucial role in emotional learning irrespective of valence. The BLA projection to the nucleus accumbens (NAc) is thought to modulate cue-triggered motivated behaviours, but our understanding of the interaction between these two brain regions has been limited by the inability to manipulate neural-circuit elements of this pathway selectively during behaviour. To circumvent this limitation, we used in vivo optogenetic stimulation or inhibition of glutamatergic fibres from the BLA to the NAc, coupled with intracranial pharmacology and ex vivo electrophysiology. Here we show that optical stimulation of the pathway from the BLA to the NAc in mice reinforces behavioural responding to earn additional optical stimulation of these synaptic inputs. Optical stimulation of these glutamatergic fibres required intra-NAc dopamine D1-type receptor signalling, but not D2-type receptor signalling. Brief optical inhibition of fibres from the BLA to the NAc reduced cue-evoked intake of sucrose, demonstrating an important role of this specific pathway in controlling naturally occurring reward-related behaviour. Moreover, although optical stimulation of glutamatergic fibres from the medial prefrontal cortex to the NAc also elicited reliable excitatory synaptic responses, optical self-stimulation behaviour was not observed by activation of this pathway. These data indicate that whereas the BLA is important for processing both positive and negative affect, the glutamatergic pathway from the BLA to the NAc, in conjunction with dopamine signalling in the NAc, promotes motivated behavioural responding. Thus, optogenetic manipulation of anatomically distinct synaptic inputs to the NAc reveals functionally distinct properties of these inputs in controlling reward-seeking behaviours.


Assuntos
Tonsila do Cerebelo/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Vias Neurais/fisiologia , Núcleo Accumbens/fisiologia , Recompensa , Tonsila do Cerebelo/citologia , Animais , Comportamento Aditivo/fisiopatologia , Channelrhodopsins , Sinais (Psicologia) , Dopamina/metabolismo , Ingestão de Líquidos , Ácido Glutâmico/metabolismo , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Nervosas/fisiologia , Neurônios/metabolismo , Núcleo Accumbens/citologia , Técnicas de Patch-Clamp , Estimulação Luminosa , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo , Sacarose/metabolismo , Sacarose/farmacologia
5.
J Neurosci ; 34(10): 3699-705, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24599468

RESUMO

Forming and breaking associations between emotionally salient environmental stimuli and rewarding or aversive outcomes is an essential component of learned adaptive behavior. Importantly, when cue-reward contingencies degrade, animals must exhibit behavioral flexibility to extinguish prior learned associations. Understanding the specific neural circuit mechanisms that operate during the formation and extinction of conditioned behaviors is critical because dysregulation of these neural processes is hypothesized to underlie many of the maladaptive and pathological behaviors observed in various neuropsychiatric disorders in humans. The medial prefrontal cortex (mPFC) participates in the behavioral adaptations seen in both appetitive and aversive-cue-mediated responding, but the precise cell types and circuit mechanisms sufficient for driving these complex behavioral states remain largely unspecified. Here, we recorded and manipulated the activity of parvalbumin-positive fast spiking interneurons (PV+ FSIs) in the prelimbic area (PrL) of the mPFC in mice. In vivo photostimulation of PV+ FSIs resulted in a net inhibition of PrL neurons, providing a circuit blueprint for behavioral manipulations. Photostimulation of mPFC PV+ cells did not alter anticipatory or consummatory licking behavior during reinforced training sessions. However, optical activation of these inhibitory interneurons to cues associated with reward significantly accelerated the extinction of behavior during non-reinforced test sessions. These data suggest that suppression of excitatory mPFC networks via increased activity of PV+ FSIs may enhance reward-related behavioral flexibility.


Assuntos
Extinção Psicológica/fisiologia , Interneurônios/metabolismo , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Recompensa , Animais , Interneurônios/citologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Estimulação Luminosa/métodos , Córtex Pré-Frontal/citologia
6.
Alcohol Clin Exp Res ; 37(10): 1680-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23763790

RESUMO

BACKGROUND: Corticotropin releasing factor (CRF) and urocortin play an important role in many stress responses and also can regulate ethanol (EtOH) intake. Adaptations in CRF signaling in the central amygdala promote EtOH consumption after long-term EtOH intake in dependent animals and also after brief periods of binge EtOH intake. Thus, even brief episodes of EtOH consumption can alter the function of the CRF system, allowing CRF to regulate EtOH intake. Here, we examined whether brief binge EtOH consumption leads to CRF receptor adaptations within the ventral tegmental area (VTA), a structure involved in signaling rewarding and aversive events and important in the development and expression of drug and alcohol addiction. METHODS: We utilized a mouse model of binge drinking known as drinking in the dark (DID), where C57BL/6J mice drink approximately 6 g/kg in 4 hours and achieve blood EtOH concentrations of approximately 100 mg/dl, which is equivalent to binge drinking in humans. We used ex vivo whole-cell recordings from putative VTA dopamine (DA) neurons to examine CRF regulation of NMDA receptor (NMDAR) currents. We also examined the impact of CRF receptor antagonist injection in the VTA on binge EtOH intake. RESULTS: Ex vivo whole-cell recordings from putative VTA DA neurons showed enhanced CRF-mediated potentiation of NMDAR currents in juvenile mice that consumed EtOH in the DID procedure. CRF-induced potentiation of NMDAR currents in EtOH-drinking mice was blocked by administration of CP-154,526 (3 µM), a selective CRF1 receptor antagonist. Furthermore, intra-VTA infusion of CP-154,526 (1 µg) significantly reduced binge EtOH consumption in adult mice. These results were not due to alterations of VTA NMDAR number or function, suggesting that binge drinking may enhance signaling through VTA CRF1 receptors onto NMDARs. CONCLUSIONS: Altered CRF1 receptor-mediated signaling in the VTA promotes binge-like EtOH consumption in mice, which supports the idea that CRF1 receptors may therefore be a promising pharmacological target for reducing binge drinking in humans.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Escuridão , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos
7.
Neuropharmacology ; 189: 108527, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33741403

RESUMO

Binge ethanol drinking is an increasingly problematic component of alcohol use disorder costing the United States approximately over $150 billion every year and causes progressive neuroplasticity alterations in numerous brain regions. However, the precise nature or machinery that underlies binge drinking has not yet been elucidated. Corticotropin releasing factor (CRF) neurons in the central amygdala (CeA) are thought to modulate binge drinking, but the specific circuit mechanisms remain poorly understood. Here, we combined optogenetics with in vivo electrophysiology to identify and record from CeA CRF neurons in mice during a repeated binge ethanol drinking task. First, we found that CeA CRF neurons were more active than CeA non-CRF cells during our binge drinking paradigm. We also observed that CeA CRF neurons displayed a heterogeneous spectrum of responses to a lick of ethanol including, pre-lick activated, lick-excited, lick-inhibited, and no response. Interestingly, pre-lick activated CeA CRF neurons exhibited higher frequency and burst firing during binge drinking sessions. Moreover, their overall tonic and phasic electrical activity enhances over repeated binge drinking sessions. Remarkably, CeA CRF units and pre-lick activated CeA CRF neurons did not show higher firing rate or bursting activity during water and sucrose consumption, suggesting that ethanol may "hijack" or plastically alter their intrinsic excitability. This article is part of the special issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Assuntos
Potenciais de Ação/fisiologia , Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Etanol/toxicidade , Neurônios/metabolismo , Potenciais de Ação/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/efeitos adversos , Consumo de Bebidas Alcoólicas/fisiopatologia , Animais , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Núcleo Central da Amígdala/efeitos dos fármacos , Núcleo Central da Amígdala/fisiopatologia , Etanol/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Microeletrodos , Neurônios/efeitos dos fármacos
8.
Neuropharmacology ; 196: 108695, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34233202

RESUMO

Modifications in brain regions that govern reward-seeking are thought to contribute to persistent behaviors that are heavily associated with alcohol-use disorder (AUD) including binge ethanol drinking. The bed nucleus of the stria terminalis (BNST) is a critical node linked to both alcohol consumption and the onset, maintenance and progression of adaptive anxiety and stress-related disorders. Differences in anatomy, connectivity and receptor subpopulations, make the BNST a sexually dimorphic region. Previous work indicates that the ventral BNST (vBNST) receives input from the insular cortex (IC), a brain region involved in processing the body's internal state. This IC-vBNST projection has also been implicated in emotional and reward-seeking processes. Therefore, we examined the functional properties of vBNST-projecting, IC neurons in male and female mice that have undergone short-term ethanol exposure and abstinence using a voluntary Drinking in the Dark paradigm (DID) paired with whole-cell slice electrophysiology. First we show that IC neurons projected predominantly to the vBNST. Next, our data show that short-term ethanol exposure and abstinence enhanced excitatory synaptic strength onto vBNST-projecting, IC neurons in both sexes. However, we observed diametrically opposing modifications in excitability across sexes. In particular, short-term ethanol exposure resulted in increased intrinsic excitability of vBNST-projecting, IC neurons in females but not in males. Furthermore, in females, abstinence decreased the excitability of these same neurons. Taken together these findings show that short-term ethanol exposure, as well as the abstinence cause sex-related adaptations in BNST-projecting, IC neurons.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Córtex Insular/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Núcleos Septais/metabolismo , Abstinência de Álcool , Animais , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Depressores do Sistema Nervoso Central/administração & dosagem , Depressores do Sistema Nervoso Central/farmacologia , Etanol/administração & dosagem , Etanol/farmacologia , Feminino , Córtex Insular/fisiopatologia , Masculino , Camundongos , Vias Neurais , Neurônios/fisiologia , Técnicas de Patch-Clamp , Núcleos Septais/fisiopatologia , Caracteres Sexuais , Fatores Sexuais
9.
Alcohol Clin Exp Res ; 34(9): 1565-73, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20586757

RESUMO

BACKGROUND: Continued consumption of alcohol despite deleterious consequences is a hallmark of alcoholism and represents a critical challenge to therapeutic intervention. Previous rat studies showed that enduring alcohol self-administration despite pairing alcohol with normally aversive stimuli was only observed after very long-term intake (>8 months). Aversion-resistant alcohol intake has been previously interpreted to indicate pathological or compulsive motivation to consume alcohol. However, given the time required to model compulsive alcohol seeking in previous studies, there is considerable interest in developing more efficient and quantitative rodent models of aversion-resistant alcohol self-administration. METHODS: Outbred Wistar rats underwent 3 to 4 months or approximately 1.5 months of intermittent, home-cage, two-bottle access (IAA) to 20% alcohol (v/v) or water. Then, after brief operant training, the effect of the bitter-tasting quinine (0.1 g/l) on the motivation to seek alcohol was quantified via progressive ratio (PR). Motivation for quinine-adulterated 2% sucrose under PR was assayed in a separate cohort of 3 to 4 months IAA rats. The effects of quinine on home-cage alcohol consumption in IAA rats and rats with continuous access to alcohol were also examined. Finally, a dose-response for quinine taste preference in IAA and continuous-access animals was determined. RESULTS: Motivation for alcohol after 3 to 4 months IAA, measured using an operant PR procedure, was not altered by adulteration of alcohol with 0.1 g/l quinine. In contrast, after 3 to 4 months of IAA, motivation for sucrose under PR was significantly reduced by adulteration of sucrose with 0.1 g/l quinine. In addition, motivation for alcohol after only approximately 1.5 months IAA was significantly reduced by adulteration of alcohol with 0.1 g/l quinine. Furthermore, home-cage alcohol intake by IAA rats was insensitive to quinine at concentrations (0.01, 0.03 g/l) that significantly reduced alcohol drinking in animals with continuous access to alcohol. Finally, no changes in quinine taste preference after 3 to 4 months IAA or continuous access to alcohol were observed. CONCLUSIONS: We have developed a novel and technically simple hybrid operant/IAA model in which quinine-resistant motivation for alcohol is evident after an experimentally tractable period of time (3 to 4 months vs. 8 months). Quinine dramatically reduced sucrose and water intake by IAA rats, indicating that continued responding for alcohol in IAA rats despite adulteration with the normally aversive quinine might reflect maladaptive or compulsive motivation for alcohol. This model could facilitate identification of novel therapeutic interventions for pathological alcohol seeking in humans.


Assuntos
Etanol/administração & dosagem , Motivação/efeitos dos fármacos , Quinina/farmacologia , Animais , Comportamento de Escolha/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Líquidos/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Autoadministração , Sacarose/administração & dosagem , Fatores de Tempo
10.
Alcohol Clin Exp Res ; 33(1): 31-42, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945225

RESUMO

BACKGROUND: The alcohol deprivation effect (ADE) is characterized by transient excessive alcohol consumption upon reinstatement of ethanol following a period of ethanol deprivation. While this phenomenon has been observed in rats using both bottle drinking (consummatory behavior) and operant self-administration (consummatory and appetitive "ethanol-seeking" behavior) procedures, ADE studies in mice have primarily relied on bottle drinking measures. Furthermore, the neurochemical pathways that modulate the ADE are not well understood. Therefore, we determined whether the ADE can be observed in C57BL/6J mice using operant self-administration procedures and if expression of the ADE is modulated by the corticotropin releasing factor-1 (CRF-1) receptor. METHODS: C57BL/6J mice were trained in a 2-hour operant self-administration paradigm to lever press for 10% ethanol or water on separate response keys. Between operant sessions, mice had access to ethanol in their homecage. Once stable responding occurred, mice were deprived of ethanol for 4 days and were then retested with ethanol in the operant paradigm for 3 consecutive days. Next, to assess the role of the CRF-1 receptor, mice were given intraperitoneal (i.p.) injection (0, 10, or 20 mg/kg) of the CRF-1 receptor antagonist CP-154,526 30 minutes before ADE testing. Additional experiments assessed (i) ADE responding in which the alternate response lever was inactive, (ii) the effects of CP-154,526 on self-administration of a 1% sucrose solution following 4 days of deprivation, and (iii) ADE responding in which mice did not received i.p. injections throughout the experiment. RESULTS: Mice exhibited a significant increase in postdeprivation lever responding for ethanol with either a water reinforced or inactive alternate lever. Interestingly, i.p. injection of a 10 mg/kg dose of CP-154,526 protected against the ADE while not affecting lever responding for a sucrose solution. Finally, baseline and deprivation-induced increases of ethanol reinforced lever responding were greater in mice not given i.p. injections. CONCLUSIONS: The ADE in C57BL/6J mice can be modeled using the operant self-administration paradigm and increased ethanol self-administration associated with the ADE is modulated by CRF-1 receptor signaling.


Assuntos
Condicionamento Operante/fisiologia , Etanol/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Transdução de Sinais/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Alcoolismo/fisiopatologia , Alcoolismo/psicologia , Animais , Condicionamento Operante/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Autoadministração , Transdução de Sinais/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/psicologia
11.
Alcohol Clin Exp Res ; 32(11): 1962-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18782340

RESUMO

BACKGROUND: Drinking in the dark (DID) procedures have recently been developed to induce high levels of ethanol drinking in C57BL/6J mice, which result in blood ethanol concentrations reaching levels that have measurable affects on physiology and/or behavior. The present study determined if increased ethanol drinking associated with DID procedures may be motivated by caloric need rather than by the postingestive pharmacological effects of ethanol. To this end, food availability was manipulated or mice were given peripheral administration of orexigenic or anorectic agents during DID procedures. METHODS: C57BL/6J had 2-hours of access to the 20% (v/v) ethanol solution beginning 3-hours into the dark cycle on days 1 to 3, and 4-hours of access to the ethanol bottle on day 4 of DID procedures. In Experiment 1, the effects of food deprivation on ethanol consumption during DID procedures was assessed. In Experiments 2 and 3, mice were given intraperitoneal (i.p.) injection of the orexigenic peptide ghrelin (0, 10 or 30 mg/kg) or the anorectic protein leptin (0 or 20 microg/g), respectively, before access to ethanol on day 4 of DID procedures. In Experiment 4, hourly consumption of food and a 0.05% saccharin solution were assessed over a period of hours that included those used with DID procedures. RESULTS: Consistent with previous research, mice achieved blood ethanol concentrations (BECs) that ranged between 100 and 150 mg% on day 4 of DID experiments. Neither food deprivation nor administration of orexigenic or anorectic compounds significantly altered ethanol drinking with DID procedures. Interestingly, mice exhibited their highest level of food and saccharin solution consumption during hours that overlapped with DID procedures. CONCLUSIONS: The present observations are inconsistent with the hypothesis that C57BL/6J mice consume large amounts of ethanol during DID procedures in order to satisfy a caloric need.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Depressores do Apetite/farmacologia , Estimulantes do Apetite/farmacologia , Privação de Alimentos/fisiologia , Alcoolismo/fisiopatologia , Animais , Depressores do Apetite/administração & dosagem , Estimulantes do Apetite/administração & dosagem , Escuridão , Modelos Animais de Doenças , Ingestão de Energia/efeitos dos fármacos , Ingestão de Energia/fisiologia , Etanol/sangue , Comportamento Alimentar/efeitos dos fármacos , Grelina/administração & dosagem , Grelina/farmacologia , Injeções Intraperitoneais , Leptina/administração & dosagem , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sacarina/metabolismo
12.
Alcohol Clin Exp Res ; 32(2): 259-65, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18162072

RESUMO

BACKGROUND: Drinking in the dark (DID) procedures have recently been developed to induce high levels of ethanol drinking in C57BL/6J mice, which result in blood ethanol concentrations (BECs) reaching levels that have measurable affects on physiology and/or behavior. The present experiments determined whether the increased ethanol drinking caused by DID procedures can be attenuated by pretreatment with CP-154,526; a corticotropin releasing factor type-1 (CRF1) receptor antagonist. METHODS: In Experiment 1, male C57BL/6J mice received ethanol (20% v/v) in place of water for 4 hours, beginning with 3 hours into the dark cycle. On the fourth day, mice were given an intraperitoneal injection of one of the 4 doses of CP-154,526 (0, 1, 3, 10 mg/kg) 30 minutes before receiving their ethanol bottle. In Experiment 2, C57BL/6J mice had 2 hours of access to the 20% ethanol solution, beginning with 3 hours into the dark cycle on days 1 to 3, and 4 hours of access to the ethanol bottle on day 4 of DID procedures. Mice were given an intraperitoneal injection of one of the 4 doses of CP-154,526 (0, 1, 3, 10 mg/kg) 30 minutes before receiving their ethanol bottle on day 4. Tail blood samples were collected immediately after the 4-hour ethanol access period on the fourth day of each experiment. Additional control experiments assessed the effects of CP-154,526 on 4-hour consumption of a 10% (w/v) sucrose solution and open-field locomotor activity. RESULTS: In Experiment 1, the vehicle-treated group consumed approximately 4.0 g/kg/4 h of ethanol and achieved BECs of approximately 30 mg%. Furthermore, pretreatment with the CRF1 receptor antagonist did not alter ethanol consumption. On the other hand, procedures used in Experiment 2 resulted in vehicle-treated mice consuming approximately 6.0 g/kg/4 h of ethanol with BECs of about 80 mg%. Additionally, the 10 mg/kg dose of CP-154,526 significantly reduced ethanol consumption and BECs to approximately 3.0 g/kg/4 h and 27 mg%, respectively, relative to vehicle-treated mice. Importantly, the 10 mg/kg dose of the CRF1R antagonist did not significantly alter 4-hour sucrose consumption or locomotor activity. CONCLUSIONS: These data indicate that CRF1R signaling modulates high, but not moderate, levels of ethanol drinking associated with DID procedures.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Aprendizagem por Associação/fisiologia , Ritmo Circadiano/fisiologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Alcoolismo/fisiopatologia , Animais , Escuridão , Relação Dose-Resposta a Droga , Etanol/sangue , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiopatologia
13.
Drug Alcohol Depend ; 90(2-3): 297-300, 2007 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-17482381

RESUMO

BACKGROUND: Neuropeptide Y (NPY) is a neuromodulator with anxiolytic properties. Recent evidence suggests that NPY modulates neurobiological responses to ethanol. Because withdrawal from ethanol is associated with elevated anxiety-like behavior, and because central NPY modulates anxiety, we assessed anxiety-like behavior in mutant mice lacking normal production of NPY (NPY-/-) and in normal wild-type mice (NPY+/+) 6h after removal of a liquid diet containing 4.5% ethanol. METHODS: NPY-/- and NPY+/+ mice on a pure 129/SvEv genetic background were given 6 days of access to a liquid ethanol diet (ED) or control diet (CD). Six hours before elevated plus maze (EPM) testing, ED was replaced with CD in the ethanol-withdrawn group. RESULTS: Ethanol-withdrawn NPY-/- mice showed significantly less open arm time and total proportion of time spent in the open arm of the EPM relative to ethanol-withdrawn NPY+/+ mice and when compared to NPY-/- and NPY+/+ mice that had access to the CD. On the other hand, ethanol-withdrawn NPY+/+ mice did not show altered EPM behavior relative to controls. CONCLUSIONS: Central NPY is protective against anxiety-like behavior stemming from exposure to and/or withdrawal from ethanol. Targets aimed at NPY receptors may be useful compounds for treating anxiety associated with ethanol dependence.


Assuntos
Transtornos de Ansiedade/induzido quimicamente , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Neuropeptídeo Y/deficiência , Neuropeptídeo Y/metabolismo , Animais , Transtornos de Ansiedade/epidemiologia , Depressores do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Mutantes
14.
ACS Chem Neurosci ; 8(2): 243-251, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984692

RESUMO

The study of neuronal ensembles in awake and behaving animals is a critical question in contemporary neuroscience research. Through the examination of calcium fluctuations, which are correlated with neuronal activity, we are able to better understand complex neural circuits. Recently, the development of technologies including two-photon microscopy, miniature microscopes, and fiber photometry has allowed us to examine calcium activity in behaving subjects over time. Visualizing changes in intracellular calcium in vivo has been accomplished utilizing GCaMP, a genetically encoded calcium indicator. GCaMP allows researchers to tag cell-type specific neurons with engineered fluorescent proteins that alter their levels of fluorescence in response to changes in intracellular calcium concentration. Even with the evolution of GCaMP, in vivo calcium imaging had yet to overcome the limitation of light scattering, which occurs when imaging from neural tissue in deep brain regions. Currently, researchers have created in vivo methods to bypass this problem; this Review will delve into three of these state of the art techniques: (1) two-photon calcium imaging, (2) single photon calcium imaging, and (3) fiber photometry. Here we discuss the advantages and disadvantages of the three techniques. Continued advances in these imaging techniques will provide researchers with unparalleled access to the inner workings of the brain.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Cálcio/metabolismo , Rede Nervosa/metabolismo , Animais , Humanos , Microscopia de Fluorescência por Excitação Multifotônica , Neurônios/metabolismo , Vigília
15.
Neuropeptides ; 38(4): 235-43, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15337375

RESUMO

In recent years, evidence has emerged suggesting that neuropeptide Y (NPY) is involved with neurobiological responses to ethanol and other drugs of abuse. Here, we provide an overview of physiological, pharmacological, and genetic research showing that: (A) administration of ethanol, as well as ethanol withdrawal, alter central NPY expression, (B) NPY modulates ethanol consumption under certain conditions, and (C) NPY signaling modulates the sedative effects of several drugs, including ethanol, sodium pentobarbital, and ketamine. Evidence suggesting possible mechanism(s) by which NPY signaling modulates ethanol consumption are considered. It is suggested that NPY may influence ethanol consumption by regulating basal levels of anxiety, by modulating the sedative effects of ethanol, and/or by modulating ethanol's rewarding properties.


Assuntos
Etanol/metabolismo , Drogas Ilícitas/metabolismo , Neuropeptídeo Y/metabolismo , Consumo de Bebidas Alcoólicas , Alcoolismo/genética , Animais , Ansiedade/metabolismo , Encéfalo/metabolismo , Humanos , Hipnóticos e Sedativos/metabolismo , Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais/fisiologia
16.
Neuropeptides ; 37(6): 321-37, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14698675

RESUMO

Ethanol is a caloric compound, and ethanol drinking and food intake are both appetitive and consummatory behaviors. Furthermore, both ethanol and food have rewarding properties. It is therefore possible that overlapping central pathways are involved with uncontrolled eating and excessive ethanol consumption. A growing list of peptides has been shown to regulate food intake and/or energy homeostasis. Peptides such as the melanocortins, corticotropin releasing factor, and cholecystokinin promote reductions of food intake while others such as galanin and neuropeptide Y stimulate feeding. The present review highlights research aimed at determining if ingestive peptides also regulate voluntary ethanol intake, with an emphasis on the melanocortins and neuropeptide Y. It is suggested that research directed at ingestive peptides may expand our understanding of the neurobiological mechanisms that drive ethanol self-administration, and may reveal new therapeutic candidates for treating alcohol abuse and alcoholism.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Alcoolismo/metabolismo , Encéfalo/fisiopatologia , Ingestão de Alimentos , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Alcoolismo/fisiopatologia , Animais , Colecistocinina/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Etanol/efeitos adversos , Galanina/metabolismo , Humanos , Entorpecentes/metabolismo , Obesidade/fisiopatologia , Receptores de Melanocortina/metabolismo , alfa-MSH/metabolismo , beta-MSH/metabolismo , gama-MSH/metabolismo
17.
Alcohol ; 30(1): 35-44, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12878273

RESUMO

Two experiments were designed to evaluate whether brief access to a saccharin-ethanol solution would function as an effective unconditioned stimulus (US) in Pavlovian-autoshaping procedures. In these experiments, the insertion of a lever conditioned stimulus (CS) was followed by the brief presentation of a sipper tube containing saccharin-ethanol US solution. Experience with this Pavlovian-autoshaping procedure engendered lever CS-directed autoshaping conditioned responses (CRs) in all rats. In Experiment 1, the concentration of ethanol [0%, 2%, 4%, 6%, or 8% (vol./vol.)] in 0.1% saccharin was systematically increased within subjects across autoshaping sessions to evaluate the relation between a rat's drinking and lever pressing. In Experiment 2, the mean intertrial interval (ITI) duration (60, 90, 120 s) was systematically increased within subjects across autoshaping sessions to evaluate the effect of ITI duration on drinking and lever pressing. A pseudoconditioning control group received lever CS randomly with respect to the saccharin-ethanol US solution. In Experiment 1, lever-press autoshaping CRs developed in all rats, and the tendency of a rat to drink an ethanol concentration was predictive of the performance of lever-press autoshaping CRs. In Experiment 2, longer ITIs induced more lever CS-directed responding, and CS-US paired procedures yielded more lever CS-directed responding than that observed in CS-US random procedures. Saccharin-ethanol is an effective US in Pavlovian-autoshaping procedures, inducing more CS-directed responding than in pseudoconditioning controls receiving CS-US random procedures. More lever CS-directed responding was observed when there was more drinking of the saccharin-ethanol US solution (Experiment 1); when the CS and US were paired, rather than random (Experiment 2); and with longer mean ITI durations (Experiment 2). This pattern of results is consistent with the hypothesis that lever CS-directed responding reflects performance of Pavlovian-autoshaping CRs.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Condicionamento Operante/efeitos dos fármacos , Etanol/farmacologia , Sacarina/farmacologia , Edulcorantes/farmacologia , Animais , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Long-Evans , Soluções
18.
Alcohol ; 31(3): 155-60, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14693264

RESUMO

Recent evidence indicates that neuropeptide Y modulates neurobiologic responses to ethanol and ethanol consumption. Resistance to the sedative effects of ethanol, voluntary ethanol consumption, or both was found to be inversely related to neuropeptide Y levels in genetically manipulated rat and mouse models. More recently, intracerebroventricular infusion of neuropeptide Y reduced ethanol drinking in rats selectively bred for high ethanol preference, but not in low-ethanol-preferring or in outbred Wistar rats. In the current study, we determined whether intracerebroventricular infusion of neuropeptide Y would reduce voluntary ethanol consumption in high-ethanol-preferring, C57BL/6 mice. We also studied ethanol-induced sedation after intracerebroventricular infusion of neuropeptide Y. Pretreatment with doses of neuropeptide Y, ranging from 3.0 to 10.0 microg, significantly augmented ethanol-induced sedation without altering locomotor activity or plasma ethanol levels. However, neither a 5.0- nor a 10.0-microg dose of neuropeptide Y altered 2-h drinking of a 10% [volume/volume (vol./vol.)] ethanol solution. Consistent with genetic evidence, the results of current pharmacologic studies provide support that neuropeptide Y modulates ethanol-induced sedation.


Assuntos
Consumo de Bebidas Alcoólicas , Etanol/administração & dosagem , Hipnóticos e Sedativos/administração & dosagem , Neuropeptídeo Y/administração & dosagem , Sono/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/prevenção & controle , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Especificidade da Espécie
19.
Neuron ; 83(3): 513-5, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25102556

RESUMO

Serotonin is an essential neuromodulator, but the precise circuit connectivity that regulates serotonergic neurons has not been well defined. Using rabies virus tracing strategies Weissbourd et al. (2014) and Pollak Dorocic et al. (2014) in this issue of Neuron and Ogawa et al. (2014) in Cell Reports provide a comprehensive map of the inputs to serotonergic neurons, highlighting the complexity and diversity of potential upstream cellular regulators.

20.
Neuropharmacology ; 76 Pt B: 320-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23752096

RESUMO

Complex motivated behavioral processes, such as those that can go awry following substance abuse and other neuropsychiatric disorders, are mediated by a distributive network of neurons that reside throughout the brain. Neural circuits within the amygdala regions, such as the basolateral amygdala (BLA), and downstream targets such as the bed nucleus of the stria terminalis (BNST), are critical neuroanatomical structures for orchestrating emotional behavioral responses that may influence motivated actions such as the reinstatement of drug seeking behavior. Here, we review the functional neurocircuitry of the BLA and the BNST, and discuss how these circuits may guide maladaptive behavioral processes such as those seen in addiction. Thus, further study of the functional connectivity within these brain regions and others may provide insight for the development of new treatment strategies for substance use disorders. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.


Assuntos
Tonsila do Cerebelo/patologia , Comportamento Aditivo/patologia , Núcleos Septais/patologia , Animais , Humanos , Vias Neurais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA