Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 583
1.
Nature ; 630(8015): 141-148, 2024 Jun.
Article En | MEDLINE | ID: mdl-38778097

Fentanyl is a powerful painkiller that elicits euphoria and positive reinforcement1. Fentanyl also leads to dependence, defined by the aversive withdrawal syndrome, which fuels negative reinforcement2,3 (that is, individuals retake the drug to avoid withdrawal). Positive and negative reinforcement maintain opioid consumption, which leads to addiction in one-fourth of users, the largest fraction for all addictive drugs4. Among the opioid receptors, µ-opioid receptors have a key role5, yet the induction loci of circuit adaptations that eventually lead to addiction remain unknown. Here we injected mice with fentanyl to acutely inhibit γ-aminobutyric acid-expressing neurons in the ventral tegmental area (VTA), causing disinhibition of dopamine neurons, which eventually increased dopamine in the nucleus accumbens. Knockdown of µ-opioid receptors in VTA abolished dopamine transients and positive reinforcement, but withdrawal remained unchanged. We identified neurons expressing µ-opioid receptors in the central amygdala (CeA) whose activity was enhanced during withdrawal. Knockdown of µ-opioid receptors in CeA eliminated aversive symptoms, suggesting that they mediate negative reinforcement. Thus, optogenetic stimulation caused place aversion, and mice readily learned to press a lever to pause optogenetic stimulation of CeA neurons that express µ-opioid receptors. Our study parses the neuronal populations that trigger positive and negative reinforcement in VTA and CeA, respectively. We lay out the circuit organization to develop interventions for reducing fentanyl addiction and facilitating rehabilitation.


Dopaminergic Neurons , Fentanyl , Nucleus Accumbens , Receptors, Opioid, mu , Reinforcement, Psychology , Substance Withdrawal Syndrome , Ventral Tegmental Area , Animals , Fentanyl/pharmacology , Receptors, Opioid, mu/metabolism , Mice , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Substance Withdrawal Syndrome/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Dopamine/metabolism , Optogenetics , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiology , Female , Mice, Inbred C57BL , Opioid-Related Disorders/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage
2.
J Neurosci Res ; 102(4): e25319, 2024 Apr.
Article En | MEDLINE | ID: mdl-38629777

The central amygdaloid nucleus (CeA) has an ancient phylogenetic development and functions relevant for animal survival. Local cells receive intrinsic amygdaloidal information that codes emotional stimuli of fear, integrate them, and send cortical and subcortical output projections that prompt rapid visceral and social behavior responses. We aimed to describe the morphology of the neurons that compose the human CeA (N = 8 adult men). Cells within CeA coronal borders were identified using the thionine staining and were further analyzed using the "single-section" Golgi method followed by open-source software procedures for two-dimensional and three-dimensional image reconstructions. Our results evidenced varied neuronal cell body features, number and thickness of primary shafts, dendritic branching patterns, and density and shape of dendritic spines. Based on these criteria, we propose the existence of 12 morphologically different spiny neurons in the human CeA and discuss the variability in the dendritic architecture within cellular types, including likely interneurons. Some dendritic shafts were long and straight, displayed few collaterals, and had planar radiation within the coronal neuropil volume. Most of the sampled neurons showed a few to moderate density of small stubby/wide spines. Long spines (thin and mushroom) were observed occasionally. These novel data address the synaptic processing and plasticity in the human CeA. Our morphological description can be combined with further transcriptomic, immunohistochemical, and electrophysiological/connectional approaches. It serves also to investigate how neurons are altered in neurological and psychiatric disorders with hindered emotional perception, in anxiety, following atrophy in schizophrenia, and along different stages of Alzheimer's disease.


Central Amygdaloid Nucleus , Male , Adult , Animals , Humans , Phylogeny , Dendritic Spines/physiology , Neurons/physiology , Interneurons
3.
Transl Psychiatry ; 14(1): 197, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38670959

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.


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
4.
Neurobiol Learn Mem ; 211: 107925, 2024 May.
Article En | MEDLINE | ID: mdl-38579895

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.


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
5.
Brain Res ; 1836: 148938, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38615924

Prepulse inhibition (PPI) of the auditory startle response, a key measure of sensorimotor gating, diminishes with age and is impaired in various neurological conditions. While PPI deficits are often associated with cognitive impairments, their reversal is routinely used in experimental systems for antipsychotic drug screening. Yet, the cellular and circuit-level mechanisms of PPI remain unclear, even under non-pathological conditions. We recently showed that brainstem neurons located in the caudal pontine reticular nucleus (PnC) expressing the glycine transporter type 2 (GlyT2±) receive inputs from the central nucleus of the amygdala (CeA) and contribute to PPI but via an uncharted pathway. Here, using tract-tracing, immunohistochemistry and in vitro optogenetic manipulations coupled to field electrophysiological recordings, we reveal the neuroanatomical distribution of GlyT2± PnC neurons and PnC-projecting CeA glutamatergic neurons and we provide mechanistic insights on how these glutamatergic inputs suppress auditory neurotransmission in PnC sections. Additionally, in vivo experiments using GlyT2-Cre mice confirm that optogenetic activation of GlyT2± PnC neurons enhances PPI and is sufficient to induce PPI in young mice, emphasizing their role. However, in older mice, PPI decline is not further influenced by inhibiting GlyT2± neurons. This study highlights the importance of GlyT2± PnC neurons in PPI and underscores their diminished activity in age-related PPI decline.


Brain Stem , Glycine Plasma Membrane Transport Proteins , Glycine , Neurons , Prepulse Inhibition , Reflex, Startle , Animals , Prepulse Inhibition/physiology , Neurons/physiology , Neurons/metabolism , Reflex, Startle/physiology , Mice , Brain Stem/physiology , Brain Stem/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Male , Glycine/metabolism , Optogenetics , Mice, Transgenic , Mice, Inbred C57BL , Synaptic Transmission/physiology , Central Amygdaloid Nucleus/physiology , Central Amygdaloid Nucleus/metabolism
6.
Mol Cell Endocrinol ; 589: 112232, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38604549

BACKGROUND: The central nucleus of the amygdala (CeA) is part of the dopaminergic reward system and controls energy balance. Recently, a cluster of neurons was identified as responsive to the orexigenic effect of ghrelin and fasting. However, the signaling pathway by which ghrelin and fasting induce feeding is unknown. AMP-activated protein kinase (AMPK) is a cellular energy sensor, and its Thr172 phosphorylation (AMPKThr172) in the mediobasal hypothalamus regulates food intake. However, whether the expression and activation of AMPK in CeA could be one of the intracellular signaling activated in response to ghrelin and fasting eliciting food intake is unknown. AIM: To evaluate the activation of AMPK into CeA in response to ghrelin, fasting, and 2-deoxy-D-glucose (2DG) and whether feeding accompanied these changes. In addition, to investigate whether the inhibition of AMPK into CeA could decrease food intake. METHODS: On a chow diet, eight-week-old Wistar male rats were stereotaxically implanted with a cannula in the CeA to inject several modulators of AMPKα1/2Thr172 phosphorylation, and we performed physiological and molecular assays. KEY FINDINGS: Fasting increased, and refeeding reduced AMPKThr172 in the CeA. Intra-CeA glucose injection decreased feeding, whereas injection of 2DG, a glucoprivation inductor, in the CeA, increased food intake and blood glucose, despite faint increases in AMPKThr172. Intra-CeA ghrelin injection increased food intake and AMPKThr172. To further confirm the role of AMPK in the CeA, chronic injection of Melanotan II (MTII) in CeA reduced body mass and food intake over seven days together with a slight decrease in AMPKThr172. SIGNIFICANCE: Our findings identified that AMPK might be part of the signaling machinery in the CeA, which responds to nutrients and hormones contributing to feeding control. The results can contribute to understanding the pathophysiological mechanisms of altered feeding behavior/consumption, such as binge eating of caloric-dense, palatable food.


AMP-Activated Protein Kinases , Central Amygdaloid Nucleus , Eating , Fasting , Ghrelin , Rats, Wistar , Animals , Male , Ghrelin/metabolism , Ghrelin/pharmacology , AMP-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Central Amygdaloid Nucleus/metabolism , Eating/drug effects , Eating/physiology , Rats , Signal Transduction/drug effects , Deoxyglucose/pharmacology , Deoxyglucose/metabolism , Feeding Behavior/drug effects , Glucose/metabolism
7.
Cells ; 13(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38667320

Neuroplasticity in the central nucleus of the amygdala (CeA) plays a key role in the modulation of pain and its aversive component. The dynorphin/kappa opioid receptor (KOR) system in the amygdala is critical for averse-affective behaviors in pain conditions, but its mechanisms are not well understood. Here, we used chemogenetic manipulations of amygdala KOR-expressing neurons to analyze the behavioral consequences in a chronic neuropathic pain model. For the chemogenetic inhibition or activation of KOR neurons in the CeA, a Cre-inducible viral vector encoding Gi-DREADD (hM4Di) or Gq-DREADD (hM3Dq) was injected stereotaxically into the right CeA of transgenic KOR-Cre mice. The chemogenetic inhibition of KOR neurons expressing hM4Di with a selective DREADD actuator (deschloroclozapine, DCZ) in sham control mice significantly decreased inhibitory transmission, resulting in a shift of inhibition/excitation balance to promote excitation and induced pain behaviors. The chemogenetic activation of KOR neurons expressing hM3Dq with DCZ in neuropathic mice significantly increased inhibitory transmission, decreased excitability, and decreased neuropathic pain behaviors. These data suggest that amygdala KOR neurons modulate pain behaviors by exerting an inhibitory tone on downstream CeA neurons. Therefore, activation of these interneurons or blockade of inhibitory KOR signaling in these neurons could restore control of amygdala output and mitigate pain.


Amygdala , Mice, Transgenic , Neuralgia , Neurons , Receptors, Opioid, kappa , Animals , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/genetics , Neuralgia/metabolism , Neuralgia/physiopathology , Neurons/metabolism , Mice , Amygdala/metabolism , Behavior, Animal , Male , Clozapine/analogs & derivatives , Clozapine/pharmacology , Central Amygdaloid Nucleus/metabolism
8.
Brain Struct Funct ; 229(5): 1179-1191, 2024 Jun.
Article En | MEDLINE | ID: mdl-38625554

It is adaptive to restrict eating under uncertainty, such as during habituation to novel foods and unfamiliar environments. However, sustained restrictive eating can become maladaptive. Currently, the neural substrates of restrictive eating are poorly understood. Using a model of feeding avoidance under novelty, our recent study identified forebrain activation patterns and found evidence that the central nucleus of the amygdala (CEA) is a core integrating node. The current study analyzed the activity of CEA inputs in male and female rats to determine if specific pathways are recruited during feeding under novelty. Recruitment of direct inputs from the paraventricular nucleus of the thalamus (PVT), the infralimbic cortex (ILA), the agranular insular cortex (AI), the hippocampal ventral field CA1, and the bed nucleus of the stria terminals (BST) was assessed with combined retrograde tract tracing and Fos induction analysis. The study found that during consumption of a novel food in a novel environment, larger number of neurons within the PVTp and the CA1 that send monosynaptic inputs to the CEA were recruited compared to controls that consumed familiar food in a familiar environment. The ILA, AI, and BST inputs to the CEA were similarly recruited across conditions. There were no sex differences in activation of any of the pathways analyzed. These results suggest that the PVTp-CEA and CA1-CEA pathways underlie feeding inhibition during novelty and could be potential sites of malfunction in excessive food avoidance.


Central Amygdaloid Nucleus , Feeding Behavior , Hippocampus , Neural Pathways , Thalamus , Animals , Male , Female , Feeding Behavior/physiology , Central Amygdaloid Nucleus/physiology , Neural Pathways/physiology , Thalamus/physiology , Hippocampus/physiology , Rats , Exploratory Behavior/physiology , Neurons/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Rats, Long-Evans
10.
Transl Psychiatry ; 14(1): 148, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38490997

Anxiety disorders affect millions of people worldwide and impair health, happiness, and productivity on a massive scale. Developmental research points to a connection between early-life behavioral inhibition and the eventual development of these disorders. Our group has previously shown that measures of behavioral inhibition in young rhesus monkeys (Macaca mulatta) predict anxiety-like behavior later in life. In recent years, clinical and basic researchers have implicated the central extended amygdala (EAc)-a neuroanatomical concept that includes the central nucleus of the amygdala (Ce) and the bed nucleus of the stria terminalis (BST)-as a key neural substrate for the expression of anxious and inhibited behavior. An improved understanding of how early-life behavioral inhibition relates to an increased lifetime risk of anxiety disorders-and how this relationship is mediated by alterations in the EAc-could lead to improved treatments and preventive strategies. In this study, we explored the relationships between infant behavioral inhibition and peri-adolescent defensive behavior and brain metabolism in 18 female rhesus monkeys. We coupled a mildly threatening behavioral assay with concurrent multimodal neuroimaging, and related those findings to various measures of infant temperament. To score the behavioral assay, we developed and validated UC-Freeze, a semi-automated machine-learning (ML) tool that uses unsupervised clustering to quantify freezing. Consistent with previous work, we found that heightened Ce metabolism predicted elevated defensive behavior (i.e., more freezing) in the presence of an unfamiliar human intruder. Although we found no link between infant-inhibited temperament and peri-adolescent EAc metabolism or defensive behavior, we did identify infant nervous temperament as a significant predictor of peri-adolescent defensive behavior. Our findings suggest a connection between infant nervous temperament and the eventual development of anxiety and depressive disorders. Moreover, our approach highlights the potential for ML tools to augment existing behavioral neuroscience methods.


Central Amygdaloid Nucleus , Humans , Animals , Female , Adolescent , Macaca mulatta , Temperament/physiology , Anxiety/metabolism , Anxiety Disorders/metabolism
11.
Cell Rep ; 43(3): 113933, 2024 Mar 26.
Article En | MEDLINE | ID: mdl-38460131

Anorexia nervosa (AN) is a serious psychiatric disease, but the neural mechanisms underlying its development are unclear. A subpopulation of amygdala neurons, marked by expression of protein kinase C-delta (PKC-δ), has previously been shown to regulate diverse anorexigenic signals. Here, we demonstrate that these neurons regulate development of activity-based anorexia (ABA), a common animal model for AN. PKC-δ neurons are located in two nuclei of the central extended amygdala (EAc): the central nucleus (CeA) and oval region of the bed nucleus of the stria terminalis (ovBNST). Simultaneous ablation of CeAPKC-δ and ovBNSTPKC-δ neurons prevents ABA, but ablating PKC-δ neurons in the CeA or ovBNST alone is not sufficient. Correspondingly, PKC-δ neurons in both nuclei show increased activity with ABA development. Our study shows how neurons in the amygdala regulate ABA by impacting both feeding and wheel activity behaviors and support a complex heterogeneous etiology of AN.


Central Amygdaloid Nucleus , Septal Nuclei , Animals , Protein Kinase C-delta/metabolism , Anorexia/metabolism , Neurons/metabolism , Central Amygdaloid Nucleus/metabolism , Neural Pathways/physiology , Septal Nuclei/physiology
12.
J Pharmacol Sci ; 154(4): 264-273, 2024 Apr.
Article En | MEDLINE | ID: mdl-38485344

The monosynaptic connection from the lateral parabrachial nucleus (LPB) to the central amygdala (CeA) serves as a fundamental pathway for transmitting nociceptive signals to the brain. The LPB receives nociceptive information from the dorsal horn and spinal trigeminal nucleus and sends it to the "nociceptive" CeA, which modulates pain-associated emotions and nociceptive sensitivity. To elucidate the role of densely expressed mu-opioid receptors (MORs) within this pathway, we investigated the effects of exogenously applied opioids on LPB-CeA synaptic transmission, employing optogenetics in mice expressing channelrhodopsin-2 in LPB neurons with calcitonin gene-related peptide (CGRP). A MOR agonist ([D-Ala2,N-Me-Phe4,Glycinol5]-enkephalin, DAMGO) significantly reduced the amplitude of light-evoked excitatory postsynaptic currents (leEPSCs), in a manner negatively correlated with an increase in the paired-pulse ratio. An antagonist of MORs significantly attenuated these effects. Notably, this antagonist significantly increased leEPSC amplitude when applied alone, an effect further amplified in mice subjected to lipopolysaccharide injection 2 h before brain isolation, yet not observed at the 24-h mark. We conclude that opioids could shut off the ascending nociceptive signal at the LPB-CeA synapse through presynaptic mechanisms. Moreover, this gating process might be modulated by endogenous opioids, and the innate immune system influences this modulation.


Calcitonin Gene-Related Peptide , Central Amygdaloid Nucleus , Mice , Animals , Calcitonin Gene-Related Peptide/metabolism , Central Amygdaloid Nucleus/metabolism , Synaptic Transmission , Neurons , Synapses/physiology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology
13.
Cell Rep ; 43(4): 113990, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38551964

The motivation to eat is suppressed by satiety and aversive stimuli such as nausea. The neural circuit mechanisms of appetite suppression by nausea are not well understood. Pkcδ neurons in the lateral subdivision of the central amygdala (CeA) suppress feeding in response to satiety signals and nausea. Here, we characterized neurons enriched in the medial subdivision (CeM) of the CeA marked by expression of Dlk1. CeADlk1 neurons are activated by nausea, but not satiety, and specifically suppress feeding induced by nausea. Artificial activation of CeADlk1 neurons suppresses drinking and social interactions, suggesting a broader function in attenuating motivational behavior. CeADlk1 neurons form projections to many brain regions and exert their anorexigenic activity by inhibition of neurons of the parabrachial nucleus. CeADlk1 neurons are inhibited by appetitive CeA neurons, but also receive long-range monosynaptic inputs from multiple brain regions. Our results illustrate a CeA circuit that regulates nausea-induced feeding suppression.


Calcium-Binding Proteins , Central Amygdaloid Nucleus , Feeding Behavior , Nausea , Neurons , Animals , Neurons/metabolism , Central Amygdaloid Nucleus/metabolism , Calcium-Binding Proteins/metabolism , Mice , Nausea/metabolism , Nausea/etiology , Male , Mice, Inbred C57BL , Intercellular Signaling Peptides and Proteins/metabolism
14.
J Physiol Sci ; 74(1): 17, 2024 Mar 12.
Article En | MEDLINE | ID: mdl-38475695

Previously, we found that serotonin (5-HT) release in the central nucleus of the amygdala (CeA) of anesthetized rats decreases in response to innocuous stroking of the skin, irrespective of stimulus laterality, but increases in response to noxious pinching applied to a hindlimb contralateral to the 5-HT measurement site. The aim of the present study was to determine whether intra-CeA 5-HT release responses to cutaneous stimulation were altered in an animal model of neuropathic pain induced by ligation of the left L5 spinal nerve. In anesthetized neuropathic pain model rats, stroking of the left hindlimb increased 5-HT release in the CeA, whereas stroking of the right hindlimb decreased it. Meanwhile, pinching of the left hindlimb increased intra-CeA 5-HT release irrespective of stimulus laterality. In conclusion, the present study demonstrated that intra-CeA 5-HT release responses to cutaneous stimulation are altered in an animal model of neuropathic pain.


Central Amygdaloid Nucleus , Neuralgia , Rats , Animals , Serotonin , Central Amygdaloid Nucleus/physiology , Spinal Nerves , Skin
15.
Behav Pharmacol ; 35(2-3): 132-146, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38451025

The nicotine acetylcholinergic receptor (nAchR) in the central nucleus of the amygdala (CeA) is known to modulate anxiety traits as well as ethanol-induced behavioral effects. Therefore, the present study investigated the role of CeA nAChR in the tolerance to ethanol anxiolysis and withdrawal-induced anxiety-related effects in rats on elevated plus maze (EPM). To develop ethanol dependence, rats were given free access to an ethanol-containing liquid diet for 10 days. To assess the development of tolerance, separate groups of rats were challenged with ethanol (2 g/kg, i.p.) on days 1, 3, 5, 7 and 10 during the period of ethanol exposure, followed by an EPM assessment. Moreover, expression of ethanol withdrawal was induced after switching ethanol-dependent rats to a liquid diet on day 11, and withdrawal-induced anxiety-like behavior was noted at different post-withdrawal time points using the EPM test. The ethanol-dependent rats were pretreated with intra-CeA (i.CeA) (bilateral) injections of nicotine (0.25 µg/rat) or mecamylamine (MEC) (5 ng/rat) before the challenge dose of ethanol on subthreshold tolerance on the 5th day or on peak tolerance day, that is, 7th or 10th, and before assessment of postwithdrawal anxiety on the 11th day on EPM. Bilateral i.CeA preadministration of nicotine before the challenge dose of ethanol on days 5, 7 and 10 exhibited enhanced tolerance, while injection of MEC, completely mitigated the tolerance to the ethanol-induced antianxiety effect. On the other hand, ethanol-withdrawn rats pretreated i.CeA with nicotine exacerbated while pretreatment with MEC, alleviated the ethanol withdrawal-induced anxiety on all time points. Thus, the present investigation indicates that stimulation of nAChR in CeA negatively modulates the ethanol-induced chronic behavioral effects on anxiety in rats. It is proposed that nAChR antagonists might be useful in the treatment of alcohol use disorder and ethanol withdrawal-related anxiety-like behavior.


Alcoholism , Central Amygdaloid Nucleus , Receptors, Nicotinic , Male , Animals , Rats , Nicotine/pharmacology , Anxiety/drug therapy , Ethanol/pharmacology
16.
J Neuroimmune Pharmacol ; 19(1): 9, 2024 Mar 02.
Article En | MEDLINE | ID: mdl-38430337

Primarily regarded as immune proteins, chemokines are emerging as a family of molecules serving neuromodulatory functions in the developing and adult brain. Among them, CXCL12 is constitutively and widely expressed in the CNS, where it was shown to act on cellular, synaptic, network, and behavioral levels. Its receptor, CXCR4, is abundant in the amygdala, a brain structure involved in pathophysiology of anxiety disorders. Dysregulation of CXCL12/CXCR4 signaling has been implicated in anxiety-related behaviors. Here we demonstrate that exogenous CXCL12 at 2 nM but not at 5 nM increased neuronal excitability in the lateral division of the rat central amygdala (CeL) which was evident in the Late-Firing but not Regular-Spiking neurons. These effects were blocked by AMD3100, a CXCR4 antagonist. Moreover, CXCL12 increased the excitability of the neurons of the basolateral amygdala (BLA) that is known to project to the CeL. However, CXCL12 increased neither the spontaneous excitatory nor spontaneous inhibitory synaptic transmission in the CeL. In summary, the data reveal specific activation of Late-Firing CeL cells along with BLA neurons by CXCL12 and suggest that this chemokine may alter information processing by the amygdala that likely contributes to anxiety and fear conditioning.


Basolateral Nuclear Complex , Central Amygdaloid Nucleus , Rats , Animals , Receptors, CXCR4/metabolism , Central Amygdaloid Nucleus/metabolism , Chemokine CXCL12/metabolism , Neurons/metabolism
17.
Nat Commun ; 15(1): 2081, 2024 Mar 07.
Article En | MEDLINE | ID: mdl-38453902

The presence of a companion can reduce fear, but the neural mechanisms underlying this social buffering of fear are incompletely known. We studied social buffering of fear in male and female, and its encoding in the amygdala of male, auditory fear-conditioned rats. Pharmacological, opto,- and/or chemogenetic interventions showed that oxytocin signaling from hypothalamus-to-central amygdala projections underlied fear reduction acutely with a companion and social buffering retention 24 h later without a companion. Single-unit recordings with optetrodes in the central amygdala revealed fear-encoding neurons (showing increased conditioned stimulus-responses after fear conditioning) inhibited by social buffering and blue light-stimulated oxytocinergic hypothalamic projections. Other central amygdala neurons showed baseline activity enhanced by blue light and companion exposure, with increased conditioned stimulus responses that persisted without the companion. Social buffering of fear thus switches the conditioned stimulus from encoding "fear" to "safety" by oxytocin-mediated recruitment of a distinct group of central amygdala "buffer neurons".


Central Amygdaloid Nucleus , Conditioning, Psychological , Rats , Male , Female , Animals , Conditioning, Psychological/physiology , Oxytocin , Rats, Wistar , Fear/physiology , Neurons
18.
Neuropsychopharmacology ; 49(7): 1151-1161, 2024 Jun.
Article En | MEDLINE | ID: mdl-38418568

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.


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
19.
J Neurochem ; 168(6): 957-960, 2024 Jun.
Article En | MEDLINE | ID: mdl-38413201

Early life stress, such as childhood abuse and neglect, is one of the major risk factors for the development of antisocial behavior. In rat models, repeated maternal separation (MS) stress, in which the pups are separated from the dams for a few hours each day during the first 2-3 weeks of life, increases aggressive behavior in adult males. This Editorial highlights an article in the current issue of the Journal of Neurochemistry that demonstrates the involvement of the central nucleus of the amygdala (CeA) in the escalation of aggressive behavior in the MS model. The authors show that MS rats exhibit higher c-Fos expression in the CeA during an aggressive encounter compared to non-isolated control rats. Unexpectedly, other amygdala subnuclei did not show differential activation between MS and control groups. Using optogenetics, they provide direct evidence that activation of CeA neurons increases intermale aggressive behavior and that bilateral CeA activation shifts behavioral patterns toward more qualitatively intense aggressive behavior than unilateral CeA activation. These findings highlight the important role of the CeA in the development of abnormal aggression and indicate that this region may be an important therapeutic target for human aggression induced by early life stress.


Aggression , Central Amygdaloid Nucleus , Maternal Deprivation , Stress, Psychological , Aggression/physiology , Aggression/psychology , Animals , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Central Amygdaloid Nucleus/metabolism , Rats , Humans , Male , Female
20.
Nature ; 625(7996): 743-749, 2024 Jan.
Article En | MEDLINE | ID: mdl-38233522

Survival requires the selection of appropriate behaviour in response to threats, and dysregulated defensive reactions are associated with psychiatric illnesses such as post-traumatic stress and panic disorder1. Threat-induced behaviours, including freezing and flight, are controlled by neuronal circuits in the central amygdala (CeA)2; however, the source of neuronal excitation of the CeA that contributes to high-intensity defensive responses is unknown. Here we used a combination of neuroanatomical mapping, in vivo calcium imaging, functional manipulations and electrophysiology to characterize a previously unknown projection from the dorsal peduncular (DP) prefrontal cortex to the CeA. DP-to-CeA neurons are glutamatergic and specifically target the medial CeA, the main amygdalar output nucleus mediating conditioned responses to threat. Using a behavioural paradigm that elicits both conditioned freezing and flight, we found that CeA-projecting DP neurons are activated by high-intensity threats in a context-dependent manner. Functional manipulations revealed that the DP-to-CeA pathway is necessary and sufficient for both avoidance behaviour and flight. Furthermore, we found that DP neurons synapse onto neurons within the medial CeA that project to midbrain flight centres. These results elucidate a non-canonical top-down pathway regulating defensive responses.


Avoidance Learning , Central Amygdaloid Nucleus , Neural Pathways , Neurons , Avoidance Learning/physiology , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/physiology , Neurons/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Excitatory Amino Acid Agents/pharmacology , Glutamic Acid/metabolism , Neural Pathways/physiology , Calcium/analysis , Electrophysiology , Pons/cytology , Pons/physiology
...