Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.438
1.
Endocrinology ; 165(5)2024 Mar 29.
Article En | MEDLINE | ID: mdl-38597659

We examined the effect of the puberty blocker, leuprolide acetate, on sex differences in juvenile rough-and-tumble play behavior and anxiety-like behavior in adolescent male and female rats. We also evaluated leuprolide treatment on gonadal and pituitary hormone levels and activity-regulated cytoskeleton-protein messenger RNA levels within the adolescent amygdala, a region important both for rough-and-tumble play and anxiety-like behavior. Our findings suggest that leuprolide treatment lowered anxiety-like behavior during adolescent development, suggesting that the maturation of gonadotropin-releasing hormone systems may be linked to increased anxiety. These data provide a potential new model to understand the emergence of increased anxiety triggered around puberty. Leuprolide also reduced masculinized levels of rough-and-tumble play behavior, lowered follicle-stimulating hormone, and produced a consistent pattern of reducing or halting sex differences of hormone levels, including testosterone, growth hormone, thyrotropin, and corticosterone levels. Therefore, leuprolide treatment not only pauses sexual development of peripheral tissues, but also reduces sex differences in hormones, brain, and behavior, allowing for better harmonization of these systems following gender-affirming hormone treatment. These data contribute to the intended use of puberty blockers in stopping sex differences from developing further with the potential benefit of lowering anxiety-like behavior.


Anxiety , Behavior, Animal , Leuprolide , Sexual Maturation , Animals , Leuprolide/pharmacology , Male , Female , Anxiety/drug therapy , Rats , Behavior, Animal/drug effects , Sexual Maturation/drug effects , Sex Characteristics , Amygdala/drug effects , Amygdala/metabolism , Corticosterone/blood , Rats, Sprague-Dawley , Testosterone/blood
2.
Eur Psychiatry ; 67(1): e33, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38572583

BACKGROUND: Amygdala subregion-based network dysfunction has been determined to be centrally implicated in major depressive disorder (MDD). Little is known about whether ketamine modulates amygdala subarea-related networks. We aimed to investigate the relationships between changes in the resting-state functional connectivity (RSFC) of amygdala subregions and ketamine treatment and to identify important neuroimaging predictors of treatment outcomes. METHODS: Thirty-nine MDD patients received six doses of ketamine (0.5 mg/kg). Depressive symptoms were assessed, and magnetic resonance imaging (MRI) scans were performed before and after treatment. Forty-five healthy controls underwent one MRI scan. Seed-to-voxel RSFC analyses were performed on the amygdala subregions, including the centromedial amygdala (CMA), laterobasal amygdala (LBA), and superficial amygdala subregions. RESULTS: Abnormal RSFC between the left LBA and the left precuneus in MDD patients is related to the therapeutic efficacy of ketamine. There were significant differences in changes in bilateral CMA RSFC with the left orbital part superior frontal gyrus and in changes in the left LBA with the right middle frontal gyrus between responders and nonresponders following ketamine treatment. Moreover, there was a difference in the RSFC of left LBA and the right superior temporal gyrus/middle temporal gyrus (STG/MTG) between responders and nonresponders at baseline, which could predict the antidepressant effect of ketamine on Day 13. CONCLUSIONS: The mechanism by which ketamine improves depressive symptoms may be related to its regulation of RSFC in the amygdala subregion. The RSFC between the left LBA and right STG/MTG may predict the response to the antidepressant effect of ketamine.


Amygdala , Antidepressive Agents , Depressive Disorder, Major , Ketamine , Magnetic Resonance Imaging , Humans , Ketamine/pharmacology , Ketamine/administration & dosage , Ketamine/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/physiopathology , Amygdala/drug effects , Amygdala/diagnostic imaging , Amygdala/physiopathology , Male , Female , Adult , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Antidepressive Agents/administration & dosage , Middle Aged , Treatment Outcome
3.
J Ethnopharmacol ; 330: 118270, 2024 Aug 10.
Article En | MEDLINE | ID: mdl-38685368

ETHNOPHARMACOLOGICAL RELEVANCE: Cheonwangbosimdan (CWBSD), a herbal medicine traditionally used for anxiety, insomnia, depression, and heart palpitations, has been reported to have anti-anxiety, antidepressant, cognitive improvement, and neuroprotective effects. AIM OF THE STUDY: The purpose of this study was to determine if CWBSD could affect post-traumatic stress disorder (PTSD)-like behaviors because it has prioritized clinical use over mechanism study. MATERIALS AND METHODS: A single prolonged stress (SPS) mouse model, a well-established animal model of PTSD, was used to investigate whether standardized CWBSD could mitigate PTSD-like behaviors through robust behavioral tests, including the elevated plus-maze test and marble burying test for measuring anxiety-like behaviors, the splash test, forced swimming test, and tail suspension test for evaluating depression-like behaviors, and the Y-maze test and novel object recognition test for assessing cognitive function. Additionally, a fear extinction test was employed to determine whether CWBSD might reverse fear memory extinction deficits. Amygdala tissue was isolated from SPS-treated mouse brain and subjected to Western blotting or quantitative PCR to explore mechanisms by which CWBSD could mitigate PTSD-like behaviors. RESULTS: CWBSD ameliorated emotional impairments and cognitive dysfunction in an SPS-induced PTSD-like mouse model. It also mitigated deficits in abnormal fear memory extinction. Protein expression levels of N-methyl-D-aspartate (NMDA) receptor subunit 2B (GluN2B) and phosphorylation levels of Ca2+/calmodulin-dependent protein kinase II in the amygdala were increased in SPS model mice and normalized by CWBSD. Additionally, co-administration of CWBSD and GluN2B-containing NMDA receptor antagonist, ifenprodil, at each sub-effective dose promoted fear memory extinction. CONCLUSIONS: CWBSD can alleviate SPS-induced PTSD-like behaviors by normalizing GluN2B-containing NMDA receptor activity in the amygdala. Therefore, CWBSD could be a promising candidate for PTSD treatment with fewer adverse effects and better efficacy than existing therapies.


Behavior, Animal , Disease Models, Animal , Receptors, N-Methyl-D-Aspartate , Stress Disorders, Post-Traumatic , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Stress Disorders, Post-Traumatic/metabolism , Male , Mice , Behavior, Animal/drug effects , Mice, Inbred C57BL , Fear/drug effects , Amygdala/drug effects , Amygdala/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Anxiety/drug therapy , Anxiety/psychology
4.
J Neuroendocrinol ; 36(5): e13384, 2024 May.
Article En | MEDLINE | ID: mdl-38516965

Psychosocial stress negatively impacts reproductive function by inhibiting pulsatile luteinizing hormone (LH) secretion. The posterodorsal medial amygdala (MePD) is responsible in part for processing stress and modulating the reproductive axis. Activation of the neurokinin 3 receptor (NK3R) suppresses the gonadotropin-releasing hormone (GnRH) pulse generator, under hypoestrogenic conditions, and NK3R activity in the amygdala has been documented to play a role in stress and anxiety. We investigate whether NK3R activation in the MePD is involved in mediating the inhibitory effect of psychosocial stress on LH pulsatility in ovariectomised female mice. First, we administered senktide, an NK3R agonist, into the MePD and monitored the effect on pulsatile LH secretion. We then delivered SB222200, a selective NK3R antagonist, intra-MePD in the presence of predator odour, 2,4,5-trimethylthiazole (TMT) and examined the effect on LH pulses. Senktide administration into the MePD dose-dependently suppresses pulsatile LH secretion. Moreover, NK3R signalling in the MePD mediates TMT-induced suppression of the GnRH pulse generator, which we verified using a mathematical model. The model verifies our experimental findings: (i) predator odour exposure inhibits LH pulses, (ii) activation of NK3R in the MePD inhibits LH pulses and (iii) NK3R antagonism in the MePD blocks stressor-induced inhibition of LH pulse frequency in the absence of ovarian steroids. These results demonstrate for the first time that NK3R neurons in the MePD mediate psychosocial stress-induced suppression of the GnRH pulse generator.


Luteinizing Hormone , Quinolines , Receptors, Neurokinin-3 , Signal Transduction , Stress, Psychological , Substance P/analogs & derivatives , Animals , Female , Receptors, Neurokinin-3/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/agonists , Luteinizing Hormone/metabolism , Stress, Psychological/metabolism , Mice , Signal Transduction/physiology , Signal Transduction/drug effects , Corticomedial Nuclear Complex/metabolism , Corticomedial Nuclear Complex/drug effects , Corticomedial Nuclear Complex/physiology , Peptide Fragments/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Mice, Inbred C57BL , Amygdala/metabolism , Amygdala/drug effects
5.
J Neurosci ; 44(17)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38438258

Acetylcholine (ACh) is released from basal forebrain cholinergic neurons in response to salient stimuli and engages brain states supporting attention and memory. These high ACh states are associated with theta oscillations, which synchronize neuronal ensembles. Theta oscillations in the basolateral amygdala (BLA) in both humans and rodents have been shown to underlie emotional memory, yet their mechanism remains unclear. Here, using brain slice electrophysiology in male and female mice, we show large ACh stimuli evoke prolonged theta oscillations in BLA local field potentials that depend upon M3 muscarinic receptor activation of cholecystokinin (CCK) interneurons (INs) without the need for external glutamate signaling. Somatostatin (SOM) INs inhibit CCK INs and are themselves inhibited by ACh, providing a functional SOM→CCK IN circuit connection gating BLA theta. Parvalbumin (PV) INs, which can drive BLA oscillations in baseline states, are not involved in the generation of ACh-induced theta, highlighting that ACh induces a cellular switch in the control of BLA oscillatory activity and establishes an internally BLA-driven theta oscillation through CCK INs. Theta activity is more readily evoked in BLA over the cortex or hippocampus, suggesting preferential activation of the BLA during high ACh states. These data reveal a SOM→CCK IN circuit in the BLA that gates internal theta oscillations and suggest a mechanism by which salient stimuli acting through ACh switch the BLA into a network state enabling emotional memory.


Acetylcholine , Cholecystokinin , Mice, Inbred C57BL , Theta Rhythm , Theta Rhythm/drug effects , Theta Rhythm/physiology , Animals , Male , Mice , Female , Acetylcholine/pharmacology , Acetylcholine/metabolism , Cholecystokinin/pharmacology , Cholecystokinin/metabolism , Interneurons/physiology , Interneurons/drug effects , Somatostatin/metabolism , Somatostatin/pharmacology , Amygdala/physiology , Amygdala/drug effects , Basolateral Nuclear Complex/physiology , Basolateral Nuclear Complex/drug effects , Nerve Net/physiology , Nerve Net/drug effects , Receptor, Muscarinic M3/physiology , Receptor, Muscarinic M3/metabolism , Parvalbumins/metabolism
6.
Phytomedicine ; 128: 155507, 2024 Jun.
Article En | MEDLINE | ID: mdl-38552430

BACKGROUND: Abnormal activation of astrocytes in the amygdala contributes to anxiety after hemorrhagic shock and resuscitation (HSR). Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB)-associated epigenetic reprogramming of astrocytic activation is crucial to anxiety. A bioactive monomer derived from Epimedium icariin (ICA) has been reported to modulate NF-κB signaling and astrocytic activation. PURPOSE: The present study aimed to investigate the effects of ICA on post-HSR anxiety disorders and its potential mechanism of action. METHODS: We first induced HSR in mice through a bleeding and re-transfusion model and selectively inhibited and activated astrocytes in the amygdala using chemogenetics. Then, ICA (40 mg/kg) was administered by oral gavage once daily for 21 days. Behavioral, electrophysiological, and pathological changes were assessed after HSR using the light-dark transition test, elevated plus maze, recording of local field potential (LFP), and immunofluorescence assays. RESULTS: Exposure to HSR reduced the duration of the light chamber and attenuated open-arm entries. Moreover, HSR exposure increased the theta oscillation power in the amygdala and upregulated NF-κB p65, H3K27ac, and H3K4me3 expression. Contrarily, chemogenetic inhibition of astrocytes significantly reversed these changes. Chemogenetic inhibition in astrocytes was simulated by ICA, but chemogenetic activation of astrocytes blocked the neuroprotective effects of ICA. CONCLUSION: ICA mitigated anxiety-like behaviors induced by HSR in mice via inhibiting astrocytic activation, which is possibly associated with NF-κB-induced epigenetic reprogramming.


Anxiety , Astrocytes , Flavonoids , Shock, Hemorrhagic , Animals , Astrocytes/drug effects , Flavonoids/pharmacology , Shock, Hemorrhagic/drug therapy , Mice , Anxiety/drug therapy , Male , Resuscitation/methods , Disease Models, Animal , Mice, Inbred C57BL , NF-kappa B/metabolism , Behavior, Animal/drug effects , Amygdala/drug effects , Epimedium/chemistry
7.
Acta Neuropsychiatr ; 36(2): 109-117, 2024 Apr.
Article En | MEDLINE | ID: mdl-36847240

OBJECTIVE: Cocaine is a highly addictive psychostimulant that affects synaptic activity with structural and functional adaptations of neurons. The transmembrane synaptic vesicle glycoprotein 2A (SV2A) of pre-synaptic vesicles is commonly used to measure synaptic density, as a novel approach to the detection of synaptic changes. We do not know if a single dose of cocaine suffices to affect pre-synaptic SV2A density, especially during adolescence when synapses undergo intense maturation. Here, we explored potential changes of pre-synaptic SV2A density in target brain areas associated with the cocaine-induced boost of dopaminergic neurotransmission, specifically testing if the effects would last after the return of dopamine levels to baseline. METHODS: We administered cocaine (20 mg/kg i.p.) or saline to rats in early adolescence, tested their activity levels and removed the brains 1 hour and 7 days after injection. To evaluate immediate and lasting effects, we did autoradiography with [3H]UCB-J, a specific tracer for SV2A, in medial prefrontal cortex, striatum, nucleus accumbens, amygdala, and dorsal and ventral areas of hippocampus. We also measured the striatal binding of [3H]GBR-12935 to test cocaine's occupancy of the dopamine transporter at both times of study. RESULTS: We found a significant increase of [3H]UCB-J binding in the dorsal and ventral sections of hippocampus 7 days after the cocaine administration compared to saline-injected rats, but no differences 1 hour after the injection. The [3H]GBR-12935 binding remained unchanged at both times. CONCLUSION: Cocaine provoked lasting changes of hippocampal synaptic SV2A density after a single exposure during adolescence.


Cocaine , Hippocampus , Membrane Glycoproteins , Animals , Rats , Amygdala/drug effects , Amygdala/metabolism , Brain/metabolism , Cocaine/metabolism , Cocaine/pharmacology , Corpus Striatum , Hippocampus/drug effects , Hippocampus/metabolism , Positron-Emission Tomography , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism
8.
Cereb Cortex ; 33(6): 2470-2484, 2023 03 10.
Article En | MEDLINE | ID: mdl-35650684

The endocannabinoid (eCB) system represents a promising neurobiological target for novel anxiolytic pharmacotherapies. Previous clinical and preclinical evidence has revealed that genetic and/or pharmacological manipulations altering eCB signaling modulate fear and anxiety behaviors. Water-insoluble eCB lipid anandamide requires chaperone proteins for its intracellular transport to degradation, a process that requires fatty acid-binding proteins (FABPs). Here, we investigated the effects of a novel FABP-5 inhibitor, SBFI-103, on fear and anxiety-related behaviors using rats. Acute intra-prelimbic cortex administration of SBFI-103 induced a dose-dependent anxiolytic response and reduced contextual fear expression. Surprisingly, both effects were reversed when a cannabinoid-2 receptor (CB2R) antagonist, AM630, was co-infused with SBFI-103. Co-infusion of the cannabinoid-1 receptor antagonist Rimonabant with SBFI-103 reversed the contextual fear response yet showed no reversal effect on anxiety. Furthermore, in vivo neuronal recordings revealed that intra-prelimbic region SBFI-103 infusion altered the activity of putative pyramidal neurons in the basolateral amygdala and ventral hippocampus, as well as oscillatory patterns within these regions in a CB2R-dependent fashion. Our findings identify a promising role for FABP5 inhibition as a potential target for anxiolytic pharmacotherapy. Furthermore, we identify a novel, CB2R-dependent FABP-5 signaling pathway in the PFC capable of strongly modulating anxiety-related behaviors and anxiety-related neuronal transmission patterns.


Anti-Anxiety Agents , Anxiety , Fatty Acid-Binding Proteins , Prefrontal Cortex , Receptor, Cannabinoid, CB2 , Animals , Rats , Amygdala/drug effects , Amygdala/metabolism , Anti-Anxiety Agents/metabolism , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/metabolism , Cannabinoids/metabolism , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Acid-Binding Proteins/metabolism , Fear/drug effects , Fear/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism
9.
Sci Rep ; 12(1): 367, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013368

The posterior pallial amygdala (PoA) is located on the basolateral caudal telencephalon, including the basal division of PoA (PoAb) and the compact division of PoA (PoAc). PoA plays a vital role in emotion regulation and is considered a part of the amygdala in birds. However, the regulatory functions responsible for motor behaviors and emotions between PoAb and PoAc are poorly understood. Therefore, we studied the structure and function of PoA by tract-tracing methods, constant current electrical stimulation, and different dopamine receptor drug injections in pigeons (Columba livia domestica). PoAb connects reciprocally with two nuclear groups in the cerebrum: 1) a continuum comprising the temporo-parieto-occipitalis, corticoidea dorsolateralis, hippocampus, and parahippocampalis areas and 2) rostral areas of the hemisphere, including the nucleus septalis lateralis and nucleus taeniae amygdalae. Extratelencephalic projections of PoAb terminate in the lateral hypothalamic nucleus and are scattered in many limbic midbrain regions. PoAb and PoAc mainly mediated the turning movement. In the 'open-field' test, D1 agonist and D2 antagonist could significantly reduce the latency period for entering into the central area and increase the residence time in the central area, whereas D1 antagonist and D2 agonist had the opposite effect. PoAb and PoAc are important brain areas that mediate turning behavior.


Amygdala/physiology , Behavior, Animal , Columbidae/physiology , Motor Activity , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/drug effects , Columbidae/metabolism , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopaminergic Neurons/physiology , Electric Stimulation , Female , Male , Motor Activity/drug effects , Neuroanatomical Tract-Tracing Techniques , Open Field Test , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism
10.
Neurobiol Dis ; 164: 105610, 2022 03.
Article En | MEDLINE | ID: mdl-34995754

Aberrant glucocorticoid signaling via glucocorticoid receptors (GR) plays a critical role in alcohol use disorder (AUD). Acute alcohol withdrawal and protracted abstinence in dependent rats are associated with increased GR signaling and changes in GR-mediated transcriptional activity in the rat central nucleus of the amygdala (CeA). The GR antagonist mifepristone decreases alcohol consumption in dependent rats during acute withdrawal and protracted abstinence. Regulation of CeA synaptic activity by GR is currently unknown. Here, we utilized mifepristone and the selective GR antagonist CORT118335 (both at 10 µM) as pharmacological tools to dissect the role of GR on GABA transmission in male, adult Sprague-Dawley rats using slice electrophysiology. We subjected rats to chronic intermittent alcohol vapor exposure for 5-7 weeks to induce alcohol dependence. A subset of dependent rats subsequently underwent protracted alcohol withdrawal for 2 weeks, and air-exposed rats served as controls. Mifepristone reduced the frequency of pharmacologically-isolated spontaneous inhibitory postsynaptic currents (sIPSC) in the CeA (medial subdivision) without affecting postsynaptic measures in all groups, suggesting decreased GABA release with the largest effect in dependent rats. CORT118335 did not significantly alter GABA transmission in naïve, but decreased sIPSC frequency in dependent rats. Similarly, mifepristone decreased amplitudes of evoked inhibitory postsynaptic potentials only in dependent rats and during protracted withdrawal. Collectively, our study provides insight into regulation of CeA GABAergic synapses by GR. Chronic ethanol enhances the efficiency of mifepristone and CORT118335, thus highlighting the potential of drugs targeting GR as a promising pharmacological avenue for the treatment of AUD.


Alcoholism/physiopathology , Amygdala/drug effects , GABAergic Neurons/drug effects , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Synapses/drug effects , Amygdala/physiopathology , Animals , GABAergic Neurons/physiology , Inhibitory Postsynaptic Potentials/drug effects , Male , Rats , Rats, Sprague-Dawley , Synapses/physiology
11.
Article En | MEDLINE | ID: mdl-34973413

Gender is considered as a pivotal determinant of mental health. Indeed, several psychiatric disorders such as anxiety and depression are more common and persistent in women than in men. In the past two decades, impaired brain energy metabolism has been highlighted as a risk factor for the development of these psychiatric disorders. However, comprehensive behavioural and neurobiological studies in brain regions relevant to anxiety and depression symptomatology are scarce. In the present study, we summarize findings describing cannabidiol effects on anxiety and depression in maternally separated female mice as a well-established rodent model of early-life stress associated with many mental disorders. Our results indicate that cannabidiol could prevent anxiolytic- and depressive-related behaviour in early-life stressed female mice. Additionally, maternal separation with early weaning (MSEW) caused long-term changes in brain oxidative metabolism in both nucleus accumbens and amygdalar complex measured by cytochrome c oxidase quantitative histochemistry. However, cannabidiol treatment could not revert brain oxidative metabolism impairment. Moreover, we identified hyperphosphorylation of mTOR and ERK 1/2 proteins in the amygdala but not in the striatum, that could also reflect altered brain intracellular signalling related with to bioenergetic impairment. Altogether, our study supports the hypothesis that MSEW induces profound long-lasting molecular changes in mTOR signalling and brain energy metabolism related to depressive-like and anxiety-like behaviours in female mice, which were partially ameliorated by CBD administration.


Anticonvulsants/administration & dosage , Anxiety/drug therapy , Cannabidiol/administration & dosage , Emotions/physiology , Maternal Deprivation , Nucleus Accumbens/drug effects , Amygdala/drug effects , Animals , Behavior, Animal , Brain/metabolism , Disease Models, Animal , Female , Mice , TOR Serine-Threonine Kinases/genetics , Weaning
12.
Alcohol Clin Exp Res ; 46(1): 66-76, 2022 01.
Article En | MEDLINE | ID: mdl-35064942

BACKGROUND: Low levels of response (low LR) to alcohol predict heavy drinking and alcohol problems. Functional magnetic resonance imaging (fMRI) studies of emotion processing have shown that low LR individuals exhibit lower activation in task-related brain regions following both placebo and alcohol administration, but these studies did not examine functional brain networks that might contribute to the phenomena. The current study expands upon the earlier results by evaluating whether functional connectivity differences between the amygdala and other brain regions modulated by emotional face processing are associated with LR. Based on prior findings, we hypothesized that low LR is related to lower functional connectivity in fronto-amygdalar functional circuits, which underlie the processing of emotional stimuli. METHODS: Secondary analyses were conducted on data from a double-blind, placebo-controlled, within-subjects, cross-over study in 108 18-to-25-year-old low and high LR sex-matched pairs without alcohol use disorder at baseline. Participants performed modified emotional faces processing tasks after receiving placebo or approximately 0.7 ml/kg of ethanol. Psychophysiological interaction analyses examined functional connectivity between left and right amygdalae and related brain circuits using LR-by-alcohol general linear models. The data included 54 sex-matched pairs with 216 fMRI scans comprising alcohol and placebo conditions. RESULTS: Compared with individuals with high LR, low LR subjects demonstrated lower functional connectivity between the amygdala and the frontal lobes, insula, and parietal regions, while processing angry and happy faces. Interactions showed lower connectivity following alcohol in low LR and higher connectivity in high LR groups. CONCLUSIONS: Low LR individuals demonstrated lower functional connectivity in response both to placebo and a modest dose of ethanol. Attenuated connectivity among low LR individuals when processing emotional faces may contribute to an impaired ability to recognize alcohol intoxication in social situations and to appraise angry and happy emotions irrespective of whether alcohol is consumed.


Amygdala/drug effects , Brain/drug effects , Emotions/physiology , Ethanol/pharmacology , Adolescent , Alcoholic Intoxication/physiopathology , Alcoholic Intoxication/psychology , Amygdala/physiopathology , Brain/physiopathology , Cross-Over Studies , Double-Blind Method , Ethanol/administration & dosage , Facial Expression , Female , Humans , Magnetic Resonance Imaging , Male , Neural Pathways/physiology , Young Adult
13.
Article En | MEDLINE | ID: mdl-34695542

BACKGROUND: Stressful episodes and high alcohol consumption during adolescence are considered major risk factors for the development of psychiatric disorders in adulthood. Identification of mechanisms underlying these early events, which enhanced vulnerability to mental illness, is essential for both their prevention and treatment. METHODS: Male Wistar rats were used to investigate the long-term effects of early restraint stress and intermittent alcohol exposure (intragastric administration of 3 g/kg ethanol; 4 days/week for 4 weeks during adolescence) on anxiety-like behavior and the expression of signaling systems associated with emotional behaviors [e.g., corticosterone, fatty acid-derived molecules and endocannabinoid enzymes, glutamate receptor subunits, corticotropin releasing hormone receptors (CRHR1 and CRHR2) and neuropeptide Y receptors (NPY1R and NPYR2)] in the blood and amygdala. RESULTS: Overall, both stress and alcohol exposure during adolescence induced anxiogenic-like behaviors, increased plasma levels of corticosterone and increases in the amygdalar expression of the cannabinoid CB2 receptor and certain subunits of glutamate receptors (i.e., mGluR1, mGluR5 and NMDAR1) in young adult rats. In addition, there were specific main effects of alcohol exposure on the expression of the cannabinoid CB1 receptor, monoacylglycerol lipase (MAGL) and NPY2R in the amygdala, and significant increases were observed in rats exposed to alcohol. Interestingly, there were significant interaction effects between restraint stress and alcohol exposure on the expression of plasma 2-arachidonoyl glycerol (2-AG), and both CRHR1,2 and NPY1R in the amygdala. Thus, the restraint stress was associated with increased 2-AG levels, which was not observed in rats exposed to alcohol. The alcohol exposure was associated with an increased expression of CRHR1,2 but the restraint stress prevented these increases (stress alcohol rats). In contrast, NPY1R was only increased in rats exposed to stress and alcohol. Finally, we did not observe any potentiation of the behavioral and molecular effects by the combination of stress and alcohol, which is concordant with an overall ceiling effect on some of the variables. CONCLUSION: Separate and combined early stress and alcohol induced a common anxious phenotype with increased corticosterone in adulthood. However, there were differences in the amygdalar expression of signaling systems involved in maladaptive changes in emotional behavior. Therefore, our results suggest the existence of partially different mechanisms for stress and alcohol exposures.


Alcoholism , Anxiety/etiology , Endocannabinoids , Glutamic Acid , Phenotype , Signal Transduction/drug effects , Stress, Physiological , Amygdala/drug effects , Animals , Male , Mental Disorders/prevention & control , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/drug effects
14.
Neurochem Int ; 152: 105224, 2022 01.
Article En | MEDLINE | ID: mdl-34798194

CRH system integrates responses to stress challenges, whereas antipsychotics may impinge on this process. Effect of haloperidol (HAL) and aripiprazole (ARI) on chronic mild stress (CMS) induced neurobehavioral and CRH/CRHR1 system changes was studied in functionally interconnected rat brain areas including prefrontal cortex (PFC), bed nucleus of the stria terminalis (BNST), hypothalamic paraventricular nucleus (PVN), hippocampus (HIP), and amygdala (AMY). Animals were exposed to CMS for 3-weeks and since the 7th day of CMS injected with vehicle (VEH), HAL (1 mg/kg) or ARI (10 mg/kg) for 4-weeks. Expression levels of CRH, CRHR1, and c-fos genes and anxiety-like and anhedonia behavioural patterns were evaluated. CMS in VEH animals suppressed CRH gene expression in the PFC and BNST, c-fos expression in all areas, except HIP, and increased CRHR1 gene expression in the HIP. Antipsychotics decreased CRH gene expression in all areas, except HIP and by CMS elevated CRHR1 expression in the HIP (ARI also in AMY). CMS and antipsychotics decreased the sucrose preference. Aripiprazole prevented CRH expression decrease in the BNST and sucrose preference induced by CMS. Haloperidol increased time spent in the EPM open arms. These data indicate that HAL and ARI selectively influenced behavioural parameters and CRH/CRHR1 gene expression levels in CMS animals.


Aripiprazole/pharmacology , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/drug effects , Haloperidol/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Animals , Antipsychotic Agents/pharmacology , Anxiety/chemically induced , Anxiety/drug therapy , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Haloperidol/metabolism , Male , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism
15.
PLoS One ; 16(12): e0260577, 2021.
Article En | MEDLINE | ID: mdl-34898621

Females are more affected by psychiatric illnesses including eating disorders, depression, and post-traumatic stress disorder than males. However, the neural mechanisms mediating these sex differences are poorly understood. Animal models can be useful in exploring such neural mechanisms. Conditioned taste aversion (CTA) is a behavioral task that assesses how animals process the competition between associated reinforcing and aversive stimuli in subsequent task performance, a process critical to healthy behavior in many domains. The purpose of the present study was to identify sex differences in this behavior and associated neural responses. We hypothesized that females would value the rewarding stimulus (Boost®) relative to the aversive stimulus (LiCl) more than males in performing CTA. We evaluated behavior (Boost® intake, LiCl-induced behaviors, ultrasonic vocalizations (USVs), CTA performance) and Fos activation in relevant brain regions after the acute stimuli [acute Boost® (AB), acute LiCl (AL)] and the context-only task control (COT), Boost® only task (BOT) and Boost®-LiCl task (BLT). Acutely, females drank more Boost® than males but showed similar aversive behaviors after LiCl. Females and males performed CTA similarly. Both sexes produced 55 kHz USVs anticipating BOT and inhibited these calls in the BLT. However, more females emitted both 22 kHz and 55 kHz USVs in the BLT than males: the latter correlated with less CTA. Estrous cycle stage also influenced 55 kHz USVs. Fos responses were similar in males and females after AB or AL. Females engaged the gustatory cortex and ventral tegmental area (VTA) more than males during the BOT and males engaged the amygdala more than females in both the BOT and BLT. Network analysis of correlated Fos responses across brain regions identified two unique networks characterizing the BOT and BLT, in both of which the VTA played a central role. In situ hybridization with RNAscope identified a population of D1-receptor expressing cells in the CeA that responded to Boost® and D2 receptor-expressing cells that responded to LiCl. The present study suggests that males and females differentially process the affective valence of a stimulus to produce the same goal-directed behavior.


Conditioning, Psychological , Proto-Oncogene Proteins c-fos/metabolism , Acoustic Stimulation , Amygdala/drug effects , Amygdala/metabolism , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Female , Lithium Chloride/pharmacology , Male , Rats , Sex Characteristics , Ultrasonics
16.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article En | MEDLINE | ID: mdl-34948457

High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin's therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin's beneficial effects.


Diet, High-Fat/adverse effects , Encephalitis/drug therapy , Metformin/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Case-Control Studies , Down-Regulation , Encephalitis/chemically induced , Encephalitis/genetics , Encephalitis/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/genetics
17.
Med Sci Monit ; 27: e933469, 2021 Oct 10.
Article En | MEDLINE | ID: mdl-34628461

BACKGROUND The aim of the present study was to investigate the potential anticonvulsant effect of methylene blue (MB) in a kainic acid (KA)-induced status epilepticus (SE) model. The effects of MB on levels of oxidative stress and glutamate (Glu) also were explored. MATERIAL AND METHODS Sixty C57BL/6 mice were randomly divided into 5 equal-sized groups: (1) controls; (2) KA; (3) MB 0.5 mg/kg+KA; (4) MB 1 mg/kg+KA; and (5) vehicle+KA. The SE model was established by intra-amygdala microinjection of KA. Behavioral observations and simultaneous electroencephalographic records of the seizures in different groups were analyzed to determine the potential anticonvulsant effect of MB. The influences of MB on oxidative stress markers and glutamate were also detected to explore the possible mechanism. RESULTS MB afforded clear protection against KA-induced acute seizure, as measured by the delayed latency of onset of generalized seizures and SE, decreased percentage of SE, and increased survival rate in mice with acute epilepsy. MB markedly increased the latency to first onset of epileptiform activity and decreased the average duration of epileptiform events, as well as the percentage of time during which the epileptiform activity occurred. Administration of MB prevented KA-induced deterioration of oxidative stress markers and Glu. CONCLUSIONS MB is protective against acute seizure in SE. This beneficial effect may be at least partially related to its potent antioxidant ability and influence on Glu level.


Antioxidants/pharmacology , Methylene Blue/pharmacology , Neuroprotective Agents/pharmacology , Status Epilepticus/prevention & control , Amygdala/drug effects , Amygdala/metabolism , Animals , Antioxidants/therapeutic use , Disease Models, Animal , Electrodes, Implanted , Electroencephalography , Glutamic Acid/analysis , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Kainic Acid/toxicity , Male , Methylene Blue/therapeutic use , Mice , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/diagnosis , Status Epilepticus/pathology
18.
Int Rev Neurobiol ; 160: 223-250, 2021.
Article En | MEDLINE | ID: mdl-34696874

Adolescence is a critical period for brain development and behavioral maturation, marked by increased risk-taking behavior and the initiation of drug use. There are significant changes in gray matter volume and pruning of synapses along with a shift in excitatory to inhibitory balance which marks the maturation of cognition and decision-making. Because of ongoing brain development, adolescents are particularly sensitive to the detrimental effects of drugs, including alcohol, which can cause long-lasting consequences into adulthood. The extended amygdala is a region critically implicated in withdrawal and negative affect such as anxiety and depression. As negative affective disorders develop during adolescence, the effects of adolescent alcohol exposure on extended amygdala circuitry needs further inquiry. Here we aim to provide a framework to discuss the existing literature on the extended amygdala, the neuroadaptations which result from alcohol use, and the intersection of factors which contribute to the long-lasting effects of this exposure.


Amygdala , Ethanol , Glutamates , Signal Transduction , Adolescent , Amygdala/drug effects , Amygdala/physiopathology , Ethanol/toxicity , Glutamates/drug effects , Glutamates/physiology , Humans , Signal Transduction/drug effects
19.
Neuropharmacology ; 198: 108779, 2021 10 15.
Article En | MEDLINE | ID: mdl-34481835

The basal amygdala (BA) has been implicated in encoding fear and its extinction. The level of serotonin (5-HT) in the BA increases due to arousal and stress related to aversive stimuli. The effects of 5-HT7 receptor (5-HT7R) activation and blockade on the activity of BA neurons have not yet been investigated. In the present study, a transgenic mouse line carrying green fluorescent protein (GFP) reporter gene was used to identify neurons that express the 5-HT7R. GFP immunoreactivity was present mainly in cells that also expressed GAD67 or parvalbumin (PV), the phenotypic markers for GABAergic interneurons. Most cells showing GFP fluorescence demonstrated firing patterns characteristic of BA inhibitory interneurons. Activation of 5-HT7Rs resulted in a depolarization and/or occurrence of spontaneous spiking activity of BA interneurons that was accompanied by an increase in the mean frequency and mean amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) recorded from BA principal neurons. These effects were blocked by a specific 5-HT7R antagonist, SB269970 and were absent in slices from 5-HT7R knockout mice. Activation of 5-HT7Rs also decreased the mean frequency of spontaneous excitatory postsynaptic currents (sEPSCs) recorded from BA principal neurons, which was blocked by the GABAA receptor antagonist picrotoxin. Neither inhibitory nor excitatory miniature postsynaptic currents (mIPSCs/mEPSCs) were affected by 5-HT7R activation. These results show that in the BA 5-HT7Rs stimulate an activity-dependent enhancement of inhibitory input from local interneurons to BA principal neurons and provide insights about the possible involvement of BA serotonergic receptors in neuronal mechanisms underlying fear memory.


Amygdala/drug effects , Neurons/drug effects , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Synapses/drug effects , Animals , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/drug effects , GABAergic Neurons/drug effects , Green Fluorescent Proteins , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenols/pharmacology , Picrotoxin/pharmacology , Receptors, GABA-A/drug effects , Receptors, Serotonin/genetics , Sulfonamides/pharmacology
20.
Int J Mol Sci ; 22(18)2021 Sep 20.
Article En | MEDLINE | ID: mdl-34576305

Neuropeptide Y (NPY) has anxiolytic-like effects and facilitates the extinction of cued and contextual fear in rodents. We previously showed that intracerebroventricular administration of NPY reduces the expression of social fear in a mouse model of social fear conditioning (SFC) and localized these effects to the dorsolateral septum (DLS) and central amygdala (CeA). In the present study, we aimed to identify the receptor subtypes that mediate these local effects of NPY. We show that NPY (0.1 nmol/0.2 µL/side) reduced the expression of SFC-induced social fear in a brain region- and receptor-specific manner in male mice. In the DLS, NPY reduced the expression of social fear by acting on Y2 receptors but not on Y1 receptors. As such, prior administration of the Y2 receptor antagonist BIIE0246 (0.2 nmol/0.2 µL/side) but not the Y1 receptor antagonist BIBO3304 trifluoroacetate (0.2 nmol/0.2 µL/side) blocked the effects of NPY in the DLS. In the CeA, however, BIBO3304 trifluoroacetate but not BIIE0246 blocked the effects of NPY, suggesting that NPY reduced the expression of social fear by acting on Y1 receptors but not Y2 receptors within the CeA. This study suggests that at least two distinct receptor subtypes are differentially recruited in the DLS and CeA to mediate the effects of NPY on the expression of social fear.


Amygdala/metabolism , Neuropeptide Y/metabolism , Phobia, Social/metabolism , Septum of Brain/metabolism , Amygdala/drug effects , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzazepines/pharmacology , Fear , Male , Mice , Phobia, Social/physiopathology , Receptors, Neuropeptide Y/metabolism , Septum of Brain/drug effects
...