Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
Neurobiol Learn Mem ; 211: 107925, 2024 May.
Article in English | MEDLINE | ID: mdl-38579895

ABSTRACT

Our previous studies found that the central amygdala (CeA) modulates cerebellum-dependent eyeblink conditioning (EBC) using muscimol inactivation. We also found that CeA inactivation decreases cerebellar neuronal activity during the conditional stimulus (CS) from the start of training. Based on these findings, we hypothesized that the CeA facilitates CS input to the cerebellum. The current study tested the CS facilitation hypothesis using optogenetic inhibition with archaerhodopsin (Arch) and excitation with channelrhodopsin (ChR2) of the CeA during EBC in male rats. Optogenetic manipulations were administered during the 400 ms tone CS or during a 400 ms pre-CS period. As predicted by the CS facilitation hypothesis CeA inhibition during the CS impaired EBC and CeA excitation during the CS facilitated EBC. Unexpectedly, CeA inhibition just prior to the CS also impaired EBC, while CeA excitation during the pre-CS pathway did not facilitate EBC. The results suggest that the CeA contributes to CS facilitation and vigilance during the pre-CS period. These putative functions of the CeA may be mediated through separate output pathways from the CeA to the cerebellum.


Subject(s)
Central Amygdaloid Nucleus , Cerebellum , Conditioning, Eyelid , Optogenetics , Animals , Male , Cerebellum/physiology , Cerebellum/drug effects , Central Amygdaloid Nucleus/physiology , Central Amygdaloid Nucleus/drug effects , Conditioning, Eyelid/physiology , Conditioning, Eyelid/drug effects , Rats , Rats, Long-Evans , Conditioning, Classical/physiology , Conditioning, Classical/drug effects
2.
Transl Psychiatry ; 14(1): 197, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670959

ABSTRACT

Alcohol use and anxiety disorders occur in both males and females, but despite sharing similar presentation and classical symptoms, the prevalence of alcohol use disorder (AUD) is lower in females. While anxiety is a symptom and comorbidity shared by both sexes, the common underlying mechanism that leads to AUD and the subsequent development of anxiety is still understudied. Using a rodent model of adolescent intermittent ethanol (AIE) exposure in both sexes, we investigated the epigenetic mechanism mediated by enhancer of zeste 2 (EZH2), a histone methyltransferase, in regulating both the expression of activity-regulated cytoskeleton-associated protein (Arc) and an anxiety-like phenotype in adulthood. Here, we report that EZH2 protein levels were significantly higher in PKC-δ positive GABAergic neurons in the central nucleus of amygdala (CeA) of adult male and female rats after AIE. Reducing protein and mRNA levels of EZH2 using siRNA infusion in the CeA prevented AIE-induced anxiety-like behavior, increased H3K27me3, decreased H3K27ac at the Arc synaptic activity response element (SARE) site, and restored deficits in Arc mRNA and protein expression in both male and female adult rats. Our data indicate that an EZH2-mediated epigenetic mechanism in the CeA plays an important role in regulating anxiety-like behavior and Arc expression after AIE in both male and female rats in adulthood. This study suggests that EZH2 may serve as a tractable drug target for the treatment of adult psychopathology after adolescent alcohol exposure.


Subject(s)
Anxiety , Central Amygdaloid Nucleus , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Ethanol , Animals , Male , Female , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Rats , Anxiety/metabolism , Anxiety/genetics , Ethanol/pharmacology , Disease Models, Animal , Alcoholism/genetics , Alcoholism/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Rats, Sprague-Dawley , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
3.
Neuropsychopharmacology ; 49(7): 1151-1161, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38418568

ABSTRACT

The central nucleus of the amygdala is known to play key roles in alcohol use and affect. Neurotensin neurons in the central nucleus of the amygdala have been shown to regulate alcohol drinking in male mice. However, little is known about which neurotransmitters released by these cells drive alcohol consumption or whether these cells drive alcohol consumption in female mice. Here we show that knockdown of GABA release from central amygdala neurotensin neurons using a Nts-cre-dependent vGAT-shRNA-based AAV strategy reduces alcohol drinking in male, but not female, mice. This manipulation did not impact avoidance behavior, except in a fasted novelty-suppressed feeding test, in which vGAT shRNA mice demonstrated increased latency to feed on a familiar high-value food reward, an effect driven by male mice. In contrast, vGAT shRNA female mice showed heightened sensitivity to thermal stimulation. These data show a role for GABA release from central amygdala neurotensin neurons in modulating consumption of rewarding substances in different motivational states.


Subject(s)
Alcohol Drinking , Central Amygdaloid Nucleus , Neurons , Neurotensin , gamma-Aminobutyric Acid , Animals , Female , Male , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Neurotensin/metabolism , gamma-Aminobutyric Acid/metabolism , Neurons/metabolism , Neurons/drug effects , Alcohol Drinking/metabolism , Alcohol Drinking/genetics , Mice , Mice, Inbred C57BL , Sex Characteristics , Ethanol/administration & dosage , Ethanol/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins
4.
Behav Brain Res ; 417: 113595, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34592375

ABSTRACT

It has been shown that kappa opioid receptor (KOR) antagonists, such as nor-binaltorphimine (nor-BNI), have antinociceptive effects in some pain models that affect the trigeminal system. Also, its anxiolytic-like effect has been extensively demonstrated in the literature. The present study aimed to investigate the systemic, local, and central effect of nor-BNI on trigeminal neuropathic pain using the infraorbital nerve constriction model (CCI-ION), as well as to evaluate its effect on anxiety-like behavior associated with this model. Animals received nor-BNI systemically; in the trigeminal ganglion (TG); in the subarachnoid space to target the spinal trigeminal nucleus caudalis (Sp5C) or in the central amygdala (CeA) 14 days after CCI-ION surgery. Systemic administration of nor-BNI caused a significant reduction of facial mechanical hyperalgesia and promoted an anxiolytic-like effect, which was detected in the elevated plus-maze and the light-dark transition tests. When administered in the TG or CeA, the KOR antagonist was able to reduce facial mechanical hyperalgesia induced by CCI-ION, but without changing the anxiety-like behavior. Moreover, no change was observed on nociception and anxiety-like behavior after nor-BNI injection into the Sp5C. The present study demonstrated antinociceptive and anxiolytic-like effects of nor-BNI in a model of trigeminal neuropathic pain. The antinociceptive effect seems to be dissociated from the anxiolytic-like effect, at both the sites involved and at the dose need to achieve the effect. In conclusion, the kappa opioid system may represent a promising target to be explored for the control of trigeminal pain and associated anxiety. However, further studies are necessary to better elucidate its functioning and modulatory role in chronic trigeminal pain states.


Subject(s)
Anxiety/drug therapy , Chronic Pain/complications , Hyperalgesia/drug therapy , Naltrexone/analogs & derivatives , Receptors, Opioid, kappa/antagonists & inhibitors , Trigeminal Neuralgia/complications , Animals , Central Amygdaloid Nucleus/drug effects , Disease Models, Animal , Male , Naltrexone/pharmacology , Nociception/drug effects , Rats , Rats, Sprague-Dawley
5.
Addict Biol ; 27(1): e13067, 2022 01.
Article in English | MEDLINE | ID: mdl-34075665

ABSTRACT

Despite strong preclinical evidence for the ability of corticotropin releasing factor 1 (CRF1) antagonists to regulate alcohol consumption, clinical trials have not yet demonstrated therapeutic effects of these compounds in alcohol use disorder (AUD) patients. Several confounding factors may limit the translation of preclinical CRF1 research to patients, including reliance on experimenter-administered alcohol instead of voluntary consumption, a preponderance of evidence collected in male subjects only and an inability to assess the effects of alcohol on specific brain circuits. A population of particular interest is the CRF1-containing neurons of the central amygdala (CeA). CRF1 CeA neurons are sensitive to ethanol, but the effects of alcohol drinking on CRF signalling within this population are unknown. In the present study, we assessed the effects of voluntary alcohol drinking on inhibitory control of CRF1+ CeA neurons from male and female CRF1:GFP mice using ex vivo electrophysiology and determined the contributions of CRF1 signalling to inhibitory control and voluntary alcohol drinking. Chronic alcohol drinking produced neuroadaptations in CRF1+ neurons that increased the sensitivity of GABAA receptor-mediated sIPSCs to the acute effects of alcohol, CRF and the CRF1 antagonist R121919, but these adaptations were more pronounced in male versus female mice. The CRF1 antagonist CP-154,526 reduced voluntary alcohol drinking in both sexes and abolished sex differences in alcohol drinking. The lack of alcohol-induced adaptation in the female CRF1 system may be related to the elevated alcohol intake exhibited by female mice and could contribute to the ineffectiveness of CRF1 antagonists in female AUD patients.


Subject(s)
Alcohol Drinking/metabolism , Central Amygdaloid Nucleus/drug effects , Neurons/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Ethanol/pharmacology , Female , Male , Mice , Pyrimidines , Pyrroles , Receptors, GABA-A , Sex Characteristics , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid
6.
Neuropharmacology ; 205: 108918, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34896402

ABSTRACT

The Central Amygdala (CeA) has been heavily implicated in many aspects of alcohol use disorder. Ethanol (EtOH) has been shown to modulate glutamatergic transmission in the lateral subdivision of the CeA, however, the exact mechanism of this modulation is still unclear. EtOH exposure is associated with increased pro-inflammatory cytokines in the CeA, and inhibition of neuroimmune cells (microglia and astrocytes) has previously been shown to reduce EtOH drinking in animal models. Since neuroimmune activation seems to be involved in many of the effects of EtOH, we hypothesized that acute EtOH exposure will increase excitatory glutamatergic transmission in the CeA via modulation of neuroimmune cells. Using ex vivo brain slice whole-cell patch clamp electrophysiology, it was found that a physiologically relevant concentration of EtOH (20 mM) significantly increased presynaptic glutamatergic transmission in the CeA. Pharmacologic and chemogenetic inhibition of astrocyte function significantly reduced the ability of EtOH to modulate CeA glutamatergic transmission with minimal impact of microglia inhibition. This finding prompted additional studies examining whether direct neuroimmune activation through lipopolysaccharide (LPS) might lead to an increase in the glutamatergic transmission in the CeA. It was found that LPS modulation of glutamatergic transmission was limited by microglia activation and required astrocyte signaling. Taken together these results support the hypothesis that acute EtOH enhances lateral CeA glutamatergic transmission through an astrocyte mediated mechanism.


Subject(s)
Astrocytes/drug effects , Central Amygdaloid Nucleus/drug effects , Central Nervous System Depressants/pharmacology , Electrophysiological Phenomena/drug effects , Ethanol/pharmacology , Glutamic Acid/drug effects , Microglia/drug effects , Animals , Mice
7.
Neuropharmacology ; 203: 108885, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34798130

ABSTRACT

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates a wide spectrum of biological processes including apoptosis, immune response and inflammation. Here, we sought to understand how S1P signaling affects neuronal excitability in the central amygdala (CeA), which is a brain region associated with fear learning, aversive memory, and the affective dimension of pain. Because the G-protein coupled S1P receptor 1 (S1PR1) has been shown to be the primary mediator of S1P signaling, we utilized S1PR1 agonist SEW2871 and S1PR1 antagonist NIBR to determine a potential role of S1PR1 in altering the cellular physiology of neurons in the lateral division of the CeA (CeL) that share the neuronal lineage marker somatostatin (Sst). CeL-Sst neurons play a critical role in expression of conditioned fear and pain modulation. Here we used transgenic breeding strategies to identify fluorescently labeled CeL-Sst neurons for electrophysiological recordings. Using principal component analysis, we identified two primary subtypes of Sst neurons within the CeL in both male and female mice. We denoted the two types regular-firing (type A) and late-firing (type B) CeL-Sst neurons. In response to SEW2871 application, Type A neurons exhibited increased input resistance, while type B neurons displayed a depolarized resting membrane potential and voltage threshold, increased current threshold, and decreased voltage height. NIBR application had no effect on CeL Sst neurons, indicating the absence of tonic S1P-induced S1PR1. Our findings reveal subtypes of Sst neurons within the CeL that are uniquely affected by S1PR1 activation, which may have implications for how S1P alters supraspinal circuits.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Membrane Potentials/physiology , Oxadiazoles/pharmacology , Somatostatin/biosynthesis , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine-1-Phosphate Receptors/metabolism , Thiophenes/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Central Amygdaloid Nucleus/drug effects , Female , Gene Expression , Male , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Somatostatin/genetics , Sphingosine-1-Phosphate Receptors/agonists
8.
Neuropharmacology ; 203: 108874, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34748860

ABSTRACT

RATIONALE: The nucleus accumbens (NAc) is important for regulating a number of behaviors, including alcohol and substance use. We previously found that chemogenetically manipulating neuronal activity in the NAc core regulates binge-like drinking in mice. The central amygdala (CeA) is also an important regulator of alcohol drinking, and projects to the NAc core. We tested whether neuronal projections from the CeA to the NAc core, or neuropeptides released by the CeA in the NAc core, could regulate binge drinking. METHODS: For experiment 1, mice were administered AAV2 Cre-GFP into the NAc core and a Cre-inducible DREADD [AAV2 DIO- hM3Dq, -hM4Di, or -mCherry control] into the CeA. We tested the effects of altering CeA to NAc core activity on binge-like ethanol intake (via "Drinking in the Dark", DID). For experiment 2, we bilaterally microinfused corticotropin releasing factor (CRF), neuropeptide Y (NPY), or somatostatin (SST) into the NAc core prior to DID. For experiment 3, we tested whether intra-NAc CRF antagonism prevented reductions in drinking induced by CNO/hM3Dq stimulation of CeA->NAc projections. RESULTS: Chemogenetically increasing activity in neurons projecting from the CeA to NAc core decreased binge-like ethanol drinking (p < 0.01). Intra-NAc core CRF mimicked chemogenetic stimulation of this pathway (p < 0.05). Binge-like drinking was unaffected by the doses of NPY and SST tested. Lastly, we found that intra-NAc CRF antagonism prevented reductions in drinking induced by chemogenetic stimulation of CeA->NAc projections. These findings demonstrate that neurons projecting from the CeA to NAc core that release CRF are capable of regulating binge-like drinking in mice.


Subject(s)
Binge Drinking/metabolism , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Nerve Net/metabolism , Nucleus Accumbens/metabolism , Animals , Central Amygdaloid Nucleus/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Female , Male , Mice , Mice, Inbred C57BL , Microinjections/methods , Nerve Net/drug effects , Neuropeptide Y/administration & dosage , Nucleus Accumbens/drug effects , Piperazines/administration & dosage
9.
Behav Brain Res ; 415: 113515, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34371088

ABSTRACT

The neuropeptide orexin-A (OX-A) has diverse functions, including maintaining arousal, autonomic control, motor activity and stress responses. These functions are regulated at different terminal regions where OX-A is released. The current study examined the physiological and behavioural effects of OX-A microinjections into the central amygdala (CeA) under basal and stressed conditions in rats. When OX-A was microinjected into the CeA and the animals returned to the home-cage, heart rate and mean arterial pressure were increased compared to vehicle-injected controls. General activity of the animal was also increased, indicating that OX-A activity in CeA contributes to increased arousal. This outcome is similar to the effects of central intracerebroventricular infusions of OX-A, as well as the cardiovascular effects previously demonstrated at many of OX's efferent hypothalamic and brainstem structures. In a second study, animals were fear-conditioned to a context by delivery of electric footshocks and then animals were re-exposed to the conditioned context at test. When OX-A was microinjected at test, freezing behaviour was reduced and there was a corresponding increase in the animal's activity but no impact on the pressor and cardiac responses (i.e, blood pressure and heart rate were unchanged). This reduction in freezing suggests that OX-A activates amygdala neurons that inhibit freezing, which is similar to the actions of other neuropeptides in the CeA that modulate the appropriate defence response to fearful stimuli. Overall, these data indicate that the CeA is an important site of OX-A modulation of cardiovascular and motor activity, as well as conditioned freezing responses.


Subject(s)
Behavior, Animal/drug effects , Blood Pressure/drug effects , Central Amygdaloid Nucleus/drug effects , Conditioning, Classical/drug effects , Fear/drug effects , Heart Rate/drug effects , Orexins/pharmacology , Animals , Male , Orexins/administration & dosage , Rats , Rats, Wistar
10.
Eur J Pharmacol ; 909: 174417, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34389313

ABSTRACT

The present study intended to investigate whether the activation of cannabinoid CB1 receptors of the ventral tegmental area (VTA), the central amygdala (CeA) and the medial prefrontal cortex (mPFC) could induce conditioned place preference or aversion (CPP or CPA) in adult male Wistar rats. The involvement of hippocampal signaling pathway of Ca2+/calmodulin-dependent protein kinase II (CaMKII)/cAMP response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) was also examined following a 3-day schedule of conditioning with the injection of arachidonylcyclopropylamide (ACPA; a selective cannabinoid CB1 receptors agonist) into the targeted sites. The results showed that intra-VTA injection of the higher dose of ACPA (5 ng/rat) caused a significant CPP associating with the increased hippocampal level of the phosphorylated (p)-CAMKII/CAMKII. Intra-mPFC injection of ACPA at 3 ng/rat caused a significant CPA associating with the decreased p-CAMKII and p-CREB levels and the increased BDNF level in the hippocampus. Moreover, intra-CeA injection of the ACPA (5 ng/rat) induced a significant CPP which was associated with the increased hippocampal levels of p-CAMKII/total (t) CAMKII, p-CREB/tCREB, and BDNF. Exposing the animals to the CPP apparatus after receiving intra-cerebral vehicle injection increased the hippocampal CAMKII/CREB/BDNF signaling pathway, confirming that CPP is an associative learning task. In all experiments, the conditioning treatment with the different doses of ACPA did not affect locomotor activity in the testing phase. Taken together, it can be concluded that cannabinoid CB1 receptors of the VTA, the CeA, and the mPFC are involved in rewarding/aversion effects through the changes in the hippocampal signaling pathways.


Subject(s)
Conditioning, Operant/drug effects , Receptor, Cannabinoid, CB1/agonists , Reward , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Models, Animal , Nerve Net/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
11.
J Psychiatry Neurosci ; 46(4): E459-E471, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34318655

ABSTRACT

Background: Orexin-A (OrxA) administration in the posterior paraventricular nucleus of the thalamus (pPVT) reinstates extinguished cocaine-seeking behaviour following extended access to the drug (a model of dependence). The pPVT receives and integrates information associated with emotionally salient events and sends excitatory inputs to brain regions involved in the expression of emotional states, such as those driving cocaine-seeking behaviour (i.e., the nucleus accumbens, the central nucleus of the amygdala [CeA], the basolateral amygdala, the bed nucleus of the stria terminalis [BNST] and the prefrontal cortex). Methods: We monitored the activation pattern of these regions (measured by Fos) during cocaine-seeking induced by OrxA administered to the pPVT. The BNST and CeA emerged as being selectively activated. To test whether the functionality of these regions was pivotal during OrxA-induced cocaine-seeking behaviour, we transiently inactivated these regions concomitantly with OrxA administration to the pPVT. We then tested the participation of corticotropin-releasing factor receptors (CRF1) in the CeA during OrxA-induced cocaine-seeking using the CRF1 antagonist CP154526. Results: We observed selective activation of the CeA and BNST during cocaine-seeking induced by OrxA administered to the pPVT, but only transient inactivation of the CeA prevented cocaine-seeking behaviour. Administration of CP154526 to the CeA prevented OrxAinduced cocaine-seeking behaviour. Limitations: The use of only male rats could have been a limitation. Other limitations could have been the use of an indirect approach to test the hypothesis that administration of OrxA to the pPVT drives cocaine-seeking via CRF1 signalling in the CeA, and a lack of analysis of the participation of CeA subregions. Conclusion: Cocaine-seeking behaviour induced by OrxA administered to the pPVT is driven by activation of the CeA via CRF1 signalling.


Subject(s)
Central Amygdaloid Nucleus/drug effects , Cocaine-Related Disorders/prevention & control , Cocaine , Orexins/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Thalamus/drug effects , Animals , Cocaine/pharmacology , Male , Orexins/administration & dosage , Rats
12.
Neuropharmacology ; 192: 108601, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33971215

ABSTRACT

The dorsolateral bed nucleus of the stria terminalis (BNSTDL) has high expression of oxytocin (OT) receptors (OTR), which were shown to facilitate cued fear. However, the role of OTR in the modulation of BNSTDL activity remains elusive. BNSTDL contains GABA-ergic neurons classified based on intrinsic membrane properties into three types. Using in vitro patch-clamp recordings in male rats, we demonstrate that OT selectively excites and increases spontaneous firing rate of Type I BNSTDL neurons. As a consequence, OT increases the frequency, but not amplitude, of spontaneous inhibitory post-synaptic currents (sIPSCs) selectively in Type II neurons, an effect abolished by OTR antagonist or tetrodotoxin, and reduces spontaneous firing rate in these neurons. These results suggest an indirect effect of OT in Type II neurons, which is mediated via OT-induced increase in firing of Type I interneurons. As Type II BNSTDL neurons were shown projecting to the central amygdala (CeA), we also recorded from retrogradely labeled BNST→CeA neurons and we show that OT increases the frequency of sIPSC in these Type II BNST→CeA output neurons. In contrast, in Type III neurons, OT reduces the amplitude, but not frequency, of both sIPSCs and evoked IPSCs via a postsynaptic mechanism without changing their intrinsic excitability. We present a model of fine-tuned modulation of BNSTDL activity by OT, which selectively excites BNSTDL interneurons and inhibits Type II BNST→CeA output neurons. These results suggest that OTR in the BNST might facilitate cued fear by inhibiting the BNST→CeA neurons.


Subject(s)
Central Amygdaloid Nucleus/drug effects , Excitatory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Oxytocin/pharmacology , Septal Nuclei/drug effects , Animals , Central Amygdaloid Nucleus/physiology , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Interneurons/physiology , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Septal Nuclei/physiology
13.
Alcohol Alcohol ; 56(5): 581-588, 2021 Aug 30.
Article in English | MEDLINE | ID: mdl-33912894

ABSTRACT

AIMS: Alcohol use disorder (AUD) is linked to hyperactivity of brain stress systems, leading to withdrawal states which drive relapse. AUD differs among the sexes, as men are more likely to have AUD than women, but women progress from casual use to binge and heavy alcohol use more quickly and are more likely to relapse into repetitive episodes of heavy drinking. In alcohol dependence animal models of AUD, the central amygdala (CeA) functions as a hub of stress and anxiety processing and gamma-Aminobutyric acid (GABA)ergic signaling within the CeA is involved in dependence-induced increases in alcohol consumption. We have shown dysregulation of CeA GABAergic synaptic signaling in alcohol dependence animal models, but previous studies have exclusively used males. METHODS: Here, we used whole-cell patch clamp electrophysiology to examine basal CeA GABAergic spontaneous inhibitory postsynaptic currents (sIPSC) and the effects of acute alcohol in both naïve and alcohol dependent rats of both sexes. RESULTS: We found that sIPSC kinetics differ between females and males, as well as between naïve and alcohol-dependent animals, with naïve females having the fastest current kinetics. Additionally, we find differences in baseline current kinetics across estrous cycle stages. In contrast to the increase in sIPSC frequency routinely found in males, acute alcohol (11-88 mM) had no effect on sIPSCs in naïve females, however the highest concentration of alcohol increased sIPSC frequency in dependent females. CONCLUSION: These results provide important insight into sex differences in CeA neuronal function and dysregulation with alcohol dependence and highlight the need for sex-specific considerations in the development of effective AUD treatment.


Subject(s)
Alcoholism/physiopathology , Central Amygdaloid Nucleus/drug effects , gamma-Aminobutyric Acid/drug effects , Animals , Ethanol/pharmacology , Female , Male , Rats , Rats, Sprague-Dawley
14.
Neuropharmacology ; 189: 108527, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741403

ABSTRACT

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'.


Subject(s)
Action Potentials/physiology , Binge Drinking/metabolism , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Ethanol/toxicity , Neurons/metabolism , Action Potentials/drug effects , Alcohol Drinking/adverse effects , Alcohol Drinking/physiopathology , Animals , Binge Drinking/physiopathology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiopathology , Ethanol/administration & dosage , Female , Male , Mice , Mice, Transgenic , Microelectrodes , Neurons/drug effects
15.
Neuropharmacology ; 189: 108530, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741404

ABSTRACT

Adolescents are phenotypically characterized with hyper-sensitivity to stress and inappropriate response to stress-inducing events. Despite behavioral distinctions from adults, investigations of developmental shifts in the function of stress peptide corticotropin-releasing factor (CRF) are generally limited. Rodent models have determined that CRF receptor 1 (CRFR1) activation within the central amygdala is associated with a stress response and induces increased GABAergic synaptic neurotransmission within adult males. To investigate age- and sex-specific function of this system, we performed whole-cell patch clamp electrophysiology in brain slices from naive adolescent (postnatal days (P) 40-49) and adult (>P70) male and female Sprague Dawley rats to assess GABAergic activity in the medial central amygdala (CeM). Our results indicate a dynamic influence of age and sex on neuronal excitability within this region, as well as basal spontaneous and miniature (m) inhibitory post-synaptic currents (IPSCs) in the CeM. In addition to replicating prior findings of CRFR1-regulated increases in mIPSC frequency in adult males, we found that the selective CRFR1 agonist, Stressin-1, attenuated mIPSC frequency in adolescent males, at a concentration that did not produce an effect in adult males. Importantly, this age-specific distinction was absent in females, as Stressin-1 attenuated mIPSC frequency in both adolescent and adult females. Finally, an increase in mIPSC frequency in response to the CRF1R antagonist, NBI 35965, was observed only in the CeM of adult males. Together, these data emphasize the robust influence of age and sex on neurophysiological function of a brain region involved in the production of the stress response.


Subject(s)
Central Amygdaloid Nucleus/metabolism , GABAergic Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/metabolism , Acenaphthenes/pharmacology , Age Factors , Animals , Central Amygdaloid Nucleus/drug effects , Female , GABAergic Neurons/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Sex Factors
16.
Int J Neuropsychopharmacol ; 24(7): 580-591, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33693669

ABSTRACT

BACKGROUND: Latent inhibition (LI) reflects an adaptive form of learning impaired in certain forms of mental illness. Glutamate receptor activity is linked to LI, but the potential role of synaptic plasticity remains unspecified. METHODS: Accordingly, the present study examined the possible role of long-term depression (LTD) in LI induced by prior exposure of rats to an auditory stimulus used subsequently as a conditional stimulus to signal a pending footshock. We employed 2 mechanistically distinct LTD inhibitors, the Tat-GluA23Y peptide that blocks endocytosis of the GluA2-containing glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, or the selective glutamate n-methyl-d-aspartate receptor 2B antagonist, Ro25-6981, administered prior to the acquisition of 2-way conditioned avoidance with or without tone pre-exposure. RESULTS: Systemic LTD blockade with the Tat-GluA23Y peptide strengthened the LI effect by further impairing acquisition of conditioned avoidance in conditional stimulus-preexposed rats compared with normal conditioning in non-preexposed controls. Systemic Ro25-6981 had no significant effects. Brain region-specific microinjections of the Tat-GluA23Y peptide into the nucleus accumbens, medial prefrontal cortex, or central or basolateral amygdala demonstrated that disruption of glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor endocytosis in the central amygdala also potentiated the LI effect. CONCLUSIONS: These data revealed a previously unknown role for central amygdala LTD in LI as a key mediator of cognitive flexibility required to respond to previously irrelevant stimuli that acquire significance through reinforcement. The findings may have relevance both for our mechanistic understanding of LI and its alteration in disease states such as schizophrenia, while further elucidating the role of LTD in learning and memory.


Subject(s)
Behavior, Animal/physiology , Cell-Penetrating Peptides/pharmacology , Central Amygdaloid Nucleus/physiology , Excitatory Amino Acid Antagonists/pharmacology , Long-Term Synaptic Depression/physiology , Neural Inhibition/physiology , Animals , Auditory Perception/drug effects , Auditory Perception/physiology , Behavior, Animal/drug effects , Central Amygdaloid Nucleus/drug effects , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Long-Term Synaptic Depression/drug effects , Male , Neural Inhibition/drug effects , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
17.
Nat Neurosci ; 24(4): 529-541, 2021 04.
Article in English | MEDLINE | ID: mdl-33589833

ABSTRACT

Oxytocin (OT) orchestrates social and emotional behaviors through modulation of neural circuits. In the central amygdala, the release of OT modulates inhibitory circuits and, thereby, suppresses fear responses and decreases anxiety levels. Using astrocyte-specific gain and loss of function and pharmacological approaches, we demonstrate that a morphologically distinct subpopulation of astrocytes expresses OT receptors and mediates anxiolytic and positive reinforcement effects of OT in the central amygdala of mice and rats. The involvement of astrocytes in OT signaling challenges the long-held dogma that OT acts exclusively on neurons and highlights astrocytes as essential components for modulation of emotional states under normal and chronic pain conditions.


Subject(s)
Astrocytes/metabolism , Central Amygdaloid Nucleus/metabolism , Emotions/physiology , Neurons/metabolism , Oxytocin/metabolism , Animals , Astrocytes/drug effects , Behavior, Animal/drug effects , Behavior, Animal/physiology , Central Amygdaloid Nucleus/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Oxytocin/pharmacology , Rats , Rats, Wistar , Receptors, Oxytocin/metabolism
18.
Addict Biol ; 26(4): e13011, 2021 07.
Article in English | MEDLINE | ID: mdl-33527681

ABSTRACT

While most individuals with access to alcohol drink it recreationally, some vulnerable individuals eventually lose control over their intake and progressively develop compulsive alcohol drinking and decreased interest in alternative sources of reinforcement, two key features of addiction. The neural and molecular mechanisms underlying this vulnerability to switch from controlled to compulsive alcohol intake have not been fully elucidated. It has been shown that rats having reduced levels of expression of the gamma-aminobutyric acid (GABA) transporter, GAT-3, in the amygdala tend to persist in seeking and drinking alcohol even when adulterated with quinine, suggesting that pharmacological interventions aimed at restoring GABA homeostasis in these individuals may provide a targeted treatment to limit compulsive alcohol drinking. Here, we tested the hypothesis that the GABAB receptor agonist baclofen, which decreases GABA release, specifically reduces compulsive alcohol drinking in vulnerable individuals. In a large cohort of Sprague-Dawley rats allowed to drink alcohol under an intermittent two-bottle choice procedure, a cluster of individuals was identified that persisted in drinking alcohol despite adulteration with quinine or when an alternative ingestive reinforcer, saccharin, was available. In these rats, which were characterized by decreased GAT-3 mRNA levels in the central amygdala, acute baclofen administration (1.5 mg/kg, intraperitoneal) resulted in a decrease in compulsive drinking. These results indicate that low GAT-3 mRNA levels in the central amygdala may represent an endophenotype of vulnerability to develop a compulsive drinking of alcohol that is shown here to be mitigated by baclofen.


Subject(s)
Alcoholism/metabolism , Baclofen/pharmacology , Polymers/metabolism , Animals , Central Amygdaloid Nucleus/drug effects , Compulsive Behavior/metabolism , Conditioning, Operant/drug effects , Ethanol/pharmacology , Male , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration
19.
J Biol Chem ; 296: 100277, 2021.
Article in English | MEDLINE | ID: mdl-33428940

ABSTRACT

Anxiety is often comorbid with pain. Delta opioid receptors (DORs) are promising targets for the treatment of pain and mental disorders with little addictive potential. However, their roles in anxiety symptoms at different stages of pain are unclear. In the current study, mice with inflammatory pain at the fourth hour following complete Freund's adjuvant (CFA) injection displayed significant anxiety-like behavior, which disappeared at the seventh day. Combining electrophysiology, optogenetics, and pharmacology, we found that activation of delta opioid receptor 1 (DOR1) in the central nucleus amygdala (CeA) inhibited both the anxiolytic excitatory input from the basolateral amygdala (BLA) and the anxiogenic excitatory input from the parabrachial nucleus (PBN). In contrast, activation of delta opioid receptor 2 (DOR2) did not affect CeA excitatory synaptic transmission in normal and 4-h CFA mice but inhibited the excitatory projection from the PBN rather than the BLA in 7-day CFA mice. Furthermore, the function of both DOR1 and DOR2 was downregulated to the point of not being detectable in the CeA of mice at the 21st day following CFA injection. Taken together, these results suggest that functional switching of DOR1 and DOR2 is associated with anxiety states at different stages of pain via modulating the activity of specific pathways (BLA-CeA and PBN-CeA).


Subject(s)
Anxiety/drug therapy , Pain/drug therapy , Receptors, Opioid, delta/genetics , Animals , Anxiety/genetics , Anxiety/pathology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/pathology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/pathology , Disease Models, Animal , Freund's Adjuvant/pharmacology , Male , Mice , Neurons/metabolism , Neurons/pathology , Optogenetics/methods , Pain/genetics , Pain/pathology , Synaptic Transmission/genetics
20.
Addict Biol ; 26(4): e12990, 2021 07.
Article in English | MEDLINE | ID: mdl-33331103

ABSTRACT

The neural adaptations that occur during the transition to alcohol dependence are not entirely understood but may include a gradual recruitment of brain stress circuitry by mesolimbic reward circuitry that is activated during early stages of alcohol use. Here, we focused on dopaminergic and nondopaminergic projections from the ventral tegmental area (VTA), important for mediating acute alcohol reinforcement, to the central nucleus of the amygdala (CeA), important for alcohol dependence-related negative affect and escalated alcohol drinking. The VTA projects directly to the CeA, but the functional relevance of this circuit is not fully established. Therefore, we combined retrograde and anterograde tracing, anatomical, and electrophysiological experiments in mice and rats to demonstrate that the CeA receives input from both dopaminergic and nondopaminergic projection neurons primarily from the lateral VTA. We then used slice electrophysiology and fos immunohistochemistry to test the effects of alcohol dependence on activity and activation profiles of CeA-projecting neurons in the VTA. Our data indicate that alcohol dependence activates midbrain projections to the central amygdala, suggesting that VTA projections may trigger plasticity in the CeA during the transition to alcohol dependence and that this circuit may be involved in mediating behavioral dysregulation associated with alcohol dependence.


Subject(s)
Alcoholism/physiopathology , Central Amygdaloid Nucleus/drug effects , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/drug effects , Male , Mice , Neural Pathways/drug effects , Rats , Reward
SELECTION OF CITATIONS
SEARCH DETAIL
...