Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 817
Filter
1.
J Cardiovasc Pharmacol ; 84(2): 227-238, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39115721

ABSTRACT

ABSTRACT: Previous studies have found that anxiety disorders may increase the incidence of atrial fibrillation (AF). More and more studies have shown that α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are involved in the occurrence and development of cardiovascular diseases. However, the role of AMPARs in AF associated with anxiety disorder remains unclear. The aim of this study was to investigate the effect of AMPARs on AF susceptibility in rats with anxiety disorder and its possible mechanism. The anxiety disorder rat model was established by unpredictable empty bottle stimulation and was treated with AMPARs agonist and antagonist. Our results showed that AMPARs antagonist treatment significantly reduced sympathetic activity, improved heart rate variability, shortened action potential duration, prolonged effective refractory period, reduced AF induction rate, and improved cardiac electrical remodeling and the expression of inflammatory factors. In addition, inhibition of AMPARs reduced the phosphorylation of IκBα and p65. Our experimental results suggest that inhibition of AMPARs can reduce autonomic remodeling, improve atrial electrical remodeling, and suppress myocardial inflammation, which provides a potential therapeutic strategy for the treatment of AF associated with anxiety disorder.


Subject(s)
Anxiety Disorders , Atrial Fibrillation , Disease Models, Animal , Heart Atria , Rats, Sprague-Dawley , Receptors, AMPA , Animals , Atrial Fibrillation/physiopathology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Male , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Atria/metabolism , Heart Atria/pathology , Receptors, AMPA/metabolism , Atrial Remodeling/drug effects , Heart Rate/drug effects , Inflammation Mediators/metabolism , Action Potentials/drug effects , Phosphorylation , Signal Transduction , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Transcription Factor RelA/metabolism , Rats , Anti-Inflammatory Agents/pharmacology , Refractory Period, Electrophysiological/drug effects , NF-KappaB Inhibitor alpha/metabolism
2.
Pharmacol Biochem Behav ; 242: 173818, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971471

ABSTRACT

Anxiety disorders are chronic, disabling psychiatric disorders, and there is a growing medical need for the development of novel pharmacotherapeutic agents showing improved efficacy and an improved side effect profile as compared with the currently prescribed anxiolytic drugs. In the course of the search for next-generation anxiolytics, neuropeptide receptors have garnered interest as potential therapeutic targets, underscored by pivotal roles in modulating stress responses and findings from animal studies using pharmacological tools. Among these neuropeptide receptors, the type 1 receptor for melanin-concentrating hormone (MCH1), which has been demonstrated to be involved in an array of physiological processes, including the regulation of stress responses and affective states, has gained attraction as a therapeutic target for drugs used in the treatment of psychiatric disorders, including anxiety disorders. To date, a plethora of MCH1 antagonists have been synthesized, and studies using MCH1 antagonists and genetically manipulated mice lacking MCH1 have revealed that the blockade of MCH1 produces anxiolytic-like effects across diverse rodent paradigms. In addition, MCH1 antagonists have been demonstrated to show a rapid onset of antidepressant-like effects; therefore, they may be effective for conditions commonly encountered in patients with anxiety disorders, which is an advantage for anxiolytic drugs. Notably, MCH1 antagonists have not manifested the undesirable side effects observed with the currently prescribed anxiolytics. All these preclinical findings testify to the potential of MCH1 antagonists as novel anxiolytics. Although there are still issues that need to be resolved prior to the initiation of clinical trials, such as elucidating the precise neuronal mechanisms underlying their anxiolytic effects and exploring pertinent biomarkers that can be used in clinical trials, MCH1 blockade appears to be an attractive way to tackle anxiety disorders.


Subject(s)
Anti-Anxiety Agents , Anxiety Disorders , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Humans , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone/metabolism , Mice
3.
Pharmacol Biochem Behav ; 244: 173842, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39069097

ABSTRACT

The gut microbiome is a vast, variable, and largely unexplored component of human biology that sits at the intersection of heritable and environmental factors, and represents a rich source of novel chemistry that is already known to be compatible with the human body. This alone would make it a promising place to search for new therapeutics, but recent work has also identified gut microbiome abnormalities in patients with a number of psychiatric disorders, including anxiety disorders-suggesting that not only treatments, but cures may lie therein. Here, we'll discuss two known "para-endogenous" anxiolytics-γ-hydroxybutyrate and the neurosteroid allopregnanolone-which have recently been discovered to be produced by the microbiome.


Subject(s)
Anti-Anxiety Agents , Gastrointestinal Microbiome , Humans , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Anxiety Disorders/drug therapy , Anxiety Disorders/microbiology , Anxiety Disorders/metabolism , Pregnanolone/therapeutic use , Pregnanolone/pharmacology , Animals , Anxiety/drug therapy , Anxiety/microbiology
4.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3373-3384, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-39041101

ABSTRACT

This study aims to explore the mechanism of Dabugan Decoction in the treatment of generalized anxiety disorder(GAD) based on network pharmacology, molecular docking, and animal experiments. Network pharmacology and molecular docking technology were used to obtain the possible targets and related signaling pathways of Dabugan Decoction in the treatment of GAD. The GAD rat model was established, and the corresponding drugs were given by gavage after randomization. After 28 days of continuous intervention, the anxiety state of rats was detected, and the pathological changes of the hippocampus were detected in each group. ELISA and Western blot were used to detect the protein expression levels of related molecules. A total of 65 drug compounds in Dabugan Decoction were obtained, involving 403 targets of action, 7 398 disease targets of GAD, and 279 common targets of "drug-disease". The key nodes in the protein-protein interaction(PPI) network were Akt1, TNF, IL-6, TP53, IL-1ß, etc. Function analysis of Gene Ontology(GO) and enrichment analysis of Kyoto Encyclopedia of Genes and Genomes(KEGG) showed that the PI3K-Akt signaling pathway was the most important pathway. The results of molecular docking showed that the core components of the drug had good binding activity with the corresponding key targets. Animal experiments showed that Dabugan Decoction could effectively improve the anxiety behavior of rats and increase the open arm end movement distance and total distance of rats in the elevated cross labyrinth, the number and stay time of entering the open box, and the time(%) and the number of entering the center of the open field. At the same time, HE staining and Nicil staining showed that the number of hippocampal nerve cells in rats increased, and they were closely arranged. The damage to the cell body was improved, and there was an increase in Nissl substances in the cells. The expression of TNF-α, IL-6, and IL-1ß in rat hippocampus decreased, and the expression of TP53, p-Akt1, and p-PI3K increased. The mechanism may be related to the activation of the PI3K-Akt signaling pathway and the inhibition of inflammatory response. Dabugan Decoction can play a good therapeutic and regulatory role in GAD, reflecting the overall effect of traditional Chinese medicine(TCM) compound and the characteristics of multiple targets and multiple pathways. At the same time, it is preliminarily discussed that the state of GAD may be improved by Dabugan Decoction via-activating PI3K-Akt signaling pathway and inhibiting inflammatory response and anti-apoptosis, thus providing experimental data support for the clinical application of Dabugan Decoction.


Subject(s)
Anxiety Disorders , Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Proto-Oncogene Proteins c-akt , Animals , Rats , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Male , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Rats, Sprague-Dawley , Signal Transduction/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Protein Interaction Maps , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Humans
5.
Nat Commun ; 15(1): 5697, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972900

ABSTRACT

Climate and environmental changes threaten human mental health, but the impacts of specific environmental conditions on neuropsychiatric disorders remain largely unclear. Here, we show the impact of a humid heat environment on the brain and the gut microbiota using a conditioned housing male mouse model. We demonstrate that a humid heat environment can cause anxiety-like behaviour in male mice. Microbial 16 S rRNA sequencing analysis reveals that a humid heat environment caused gut microbiota dysbiosis (e.g., decreased abundance of Lactobacillus murinus), and metabolomics reveals an increase in serum levels of secondary bile acids (e.g., lithocholic acid). Moreover, increased neuroinflammation is indicated by the elevated expression of proinflammatory cytokines in the serum and cortex, activated PI3K/AKT/NF-κB signalling and a microglial response in the cortex. Strikingly, transplantation of the microbiota from mice reared in a humid heat environment readily recapitulates these abnormalities in germ-free mice, and these abnormalities are markedly reversed by Lactobacillus murinus administration. Human samples collected during the humid heat season also show a decrease in Lactobacillus murinus abundance and an increase in the serum lithocholic acid concentration. In conclusion, gut microbiota dysbiosis induced by a humid heat environment drives the progression of anxiety disorders by impairing bile acid metabolism and enhancing neuroinflammation, and probiotic administration is a potential therapeutic strategy for these disorders.


Subject(s)
Anxiety , Bile Acids and Salts , Dysbiosis , Gastrointestinal Microbiome , Hot Temperature , Animals , Male , Mice , Bile Acids and Salts/metabolism , Humans , Dysbiosis/microbiology , Anxiety/microbiology , Mice, Inbred C57BL , Humidity , Lithocholic Acid/metabolism , Lactobacillus , Brain/metabolism , NF-kappa B/metabolism , RNA, Ribosomal, 16S/genetics , Disease Models, Animal , Anxiety Disorders/metabolism , Anxiety Disorders/microbiology , Anxiety Disorders/etiology , Signal Transduction , Cytokines/metabolism
6.
Pharmacol Biochem Behav ; 242: 173817, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002806

ABSTRACT

Most cases of anxiety are currently treated with either benzodiazepines or serotonin reuptake inhibitors. These drugs carry with them risks for a multitude of side effects, and patient compliance suffers for this reason. There is thus a need for novel anxiolytics, and among the most compelling prospects in this vein is the study of the TAARs. The anxiolytic potential of ulotaront, a full agonist at the human TAAR1, is currently being investigated in patients with generalized anxiety disorder. Irrespective of whether this compound succeeds in clinical trials, a growing body of preclinical literature underscores the relevance of modulating the TAARs in anxiety. Multiple behavioral paradigms show anxiolytic-like effects in rodents, possibly due to increased neurogenesis and plasticity, in addition to a panoply of interactions between the TAARs and other systems. Crucially, multiple lines of evidence suggest that the TAARs, particularly TAAR1, TAAR2, and TAAR5, are expressed in the extended amygdala and hippocampus. These regions are central in the actuation of anxiety, and are particularly susceptible to neurogenic and neuroplastic effects which the TAARs are now known to regulate. The TAARs also regulate the dopamine and serotonin systems, both of which are implicated in anxiety. Ligands of the TAARs may thus constitute a new class of anxiolytics.


Subject(s)
Anti-Anxiety Agents , Receptors, G-Protein-Coupled , Humans , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Animals , Ligands , Receptors, G-Protein-Coupled/metabolism , Anxiety/drug therapy , Anxiety/metabolism , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Oxazoles/pharmacology , Trace Amine-Associated Receptors
7.
Drug Des Devel Ther ; 18: 2143-2167, 2024.
Article in English | MEDLINE | ID: mdl-38882045

ABSTRACT

Over the past decade, the idea of targeting the endocannabinoid system to treat anxiety disorders has received increasing attention. Previous studies focused more on developing cannabinoid receptor agonists or supplementing exogenous cannabinoids, which are prone to various adverse effects due to their strong pharmacological activity and poor receptor selectivity, limiting their application in clinical research. Endocannabinoid hydrolase inhibitors are considered to be the most promising development strategies for the treatment of anxiety disorders. More recent efforts have emphasized that inhibition of two major endogenous cannabinoid hydrolases, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), indirectly activates cannabinoid receptors by increasing endogenous cannabinoid levels in the synaptic gap, circumventing receptor desensitization resulting from direct enhancement of endogenous cannabinoid signaling. In this review, we comprehensively summarize the anxiolytic effects of MAGL and FAAH inhibitors and their potential pharmacological mechanisms, highlight reported novel inhibitors or natural products, and provide an outlook on future directions in this field.


Subject(s)
Amidohydrolases , Anti-Anxiety Agents , Endocannabinoids , Enzyme Inhibitors , Monoacylglycerol Lipases , Humans , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/chemistry , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , Animals , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism
8.
Biochem Biophys Res Commun ; 724: 150218, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38865810

ABSTRACT

Evidence indicates that anxiety disorders arise from an imbalance in the functioning of brain circuits that govern the modulation of emotional responses to possibly threatening stimuli. The circuits under consideration in this context include the amygdala's bottom-up activity, which signifies the existence of stimuli that may be seen as dangerous. Moreover, these circuits encompass top-down regulatory processes that originate in the prefrontal cortex, facilitating the communication of the emotional significance associated with the inputs. Diverse databases (e.g., Pubmed, ScienceDirect, Web of Science, Google Scholar) were searched for literature using a combination of different terms e.g., "anxiety", "stress", "neuroanatomy", and "neural circuits", etc. A decrease in GABAergic activity is present in both anxiety disorders and severe depression. Research on cerebral functional imaging in depressive individuals has shown reduced levels of GABA within the cortical regions. Additionally, animal studies demonstrated that a reduction in the expression of GABAA/B receptors results in a behavioral pattern resembling anxiety. The amygdala consists of inhibitory networks composed of GABAergic interneurons, responsible for modulating anxiety responses in both normal and pathological conditions. The GABAA receptor has allosteric sites (e.g., α/γ, γ/ß, and α/ß) which enable regulation of neuronal inhibition in the amygdala. These sites serve as molecular targets for anxiolytic medications such as benzodiazepine and barbiturates. Alterations in the levels of naturally occurring regulators of these allosteric sites, along with alterations to the composition of the GABAA receptor subunits, could potentially act as mechanisms via which the extent of neuronal inhibition is diminished in pathological anxiety disorders.


Subject(s)
Amygdala , Anxiety Disorders , gamma-Aminobutyric Acid , Humans , Animals , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , gamma-Aminobutyric Acid/metabolism , Amygdala/metabolism , Amygdala/physiopathology , Anxiety/metabolism , Anxiety/physiopathology , Receptors, GABA-A/metabolism
9.
Neurosci Biobehav Rev ; 163: 105741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838875

ABSTRACT

Anxiety disorders constitute a spectrum of psychological conditions affecting millions of individuals worldwide, imposing a significant health burden. Historically, the development of anxiolytic medications has been largely focused on neurotransmitter function and modulation. However, in recent years, neurolipids emerged as a prime target for understanding psychiatric pathogenesis and developing novel medications. Neurolipids influence various neural activities such as neurotransmission and cellular functioning, as well as maintaining cell membrane integrity. Therefore, this review aims to elucidate the alterations in neurolipids associated with an anxious mental state and explore their potential as targets of novel anxiolytic medications. Existing evidence tentatively associates dysregulated neurolipid levels with the etiopathology of anxiety disorders. Notably, preclinical investigations suggest that several neurolipids, including endocannabinoids and polyunsaturated fatty acids, may hold promise as potential pharmacological targets. Overall, the current literature tentatively suggests the involvement of lipids in the pathogenesis of anxiety disorders, hinting at potential prospects for future pharmacological interventions.


Subject(s)
Anxiety Disorders , Humans , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Animals , Endocannabinoids/metabolism , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Lipid Metabolism/physiology , Lipid Metabolism/drug effects
10.
J Affect Disord ; 358: 129-137, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38697224

ABSTRACT

BACKGROUND: The underlying pathogenesis of anxiety remain elusive, making the pinpointing of potential therapeutic and diagnostic biomarkers for anxiety paramount to its efficient treatment. METHODS: We undertook a proteome-wide association study (PWAS), fusing human brain proteomes from both discovery (ROS/MAP; N = 376) and validation cohorts (Banner; N = 152) with anxiety genome-wide association study (GWAS) summary statistics. Complementing this, we executed transcriptome-wide association studies (TWAS) leveraging human brain transcriptomic data from the Common Mind Consortium (CMC) to discern the confluence of genetic influences spanning both proteomic and transcriptomic levels. We further scrutinized significant genes through a suite of methodologies. RESULTS: We discerned 14 genes instrumental in the genesis of anxiety through their specific cis-regulated brain protein abundance. Out of these, 6 were corroborated in the confirmatory PWAS, with 4 also showing associations with anxiety via their cis-regulated brain mRNA levels. A heightened confidence level was attributed to 5 genes (RAB27B, CCDC92, BTN2A1, TMEM106B, and DOC2A), taking into account corroborative evidence from both the confirmatory PWAS and TWAS, coupled with insights from mendelian randomization analysis and colocalization evaluations. A majority of the identified genes manifest in brain regions intricately linked to anxiety and predominantly partake in lysosomal metabolic processes. LIMITATIONS: The limited scope of the brain proteome reference datasets, stemming from a relatively modest sample size, potentially curtails our grasp on the entire gamut of genetic effects. CONCLUSION: The genes pinpointed in our research present a promising groundwork for crafting therapeutic interventions and diagnostic tools for anxiety.


Subject(s)
Anxiety , Brain , Genome-Wide Association Study , Proteome , Humans , Proteome/genetics , Brain/metabolism , Anxiety/genetics , Anxiety/metabolism , Transcriptome , Proteomics , Anxiety Disorders/genetics , Anxiety Disorders/metabolism
11.
Int J Psychophysiol ; 200: 112353, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641018

ABSTRACT

The link between neuroticism and the various indicators of daily cortisol fluctuations is frequently noted to be inconsistent or lacking in strength. The current study aimed to investigate the predictive capacity of both self-assessment and external evaluations of neuroticism, along with their interaction, on multiple indices of diurnal cortisol variations. This research involved the assessment of neuroticism using self-report and external evaluations among 166 working individuals, coupled with the collection of saliva samples over two consecutive workdays. Employing multilevel response surface analysis, our findings indicated that self-reported neuroticism exhibited a stronger association with cortisol indices compared to external evaluations. Additionally, the level of alignment between self-assessment and external ratings of neuroticism specifically impacted the prediction of estimates of daily cortisol production. We discuss the theoretical and practical implications of these results.


Subject(s)
Circadian Rhythm , Hydrocortisone , Neuroticism , Saliva , Humans , Hydrocortisone/metabolism , Neuroticism/physiology , Male , Female , Adult , Saliva/chemistry , Saliva/metabolism , Circadian Rhythm/physiology , Young Adult , Anxiety Disorders/metabolism , Self Report , Middle Aged , Self-Assessment
12.
J Neurosci ; 44(19)2024 May 08.
Article in English | MEDLINE | ID: mdl-38569927

ABSTRACT

GPR37L1 is an orphan receptor that couples through heterotrimeric G-proteins to regulate physiological functions. Since its role in humans is not fully defined, we used an unbiased computational approach to assess the clinical significance of rare G-protein-coupled receptor 37-like 1 (GPR37L1) genetic variants found among 51,289 whole-exome sequences from the DiscovEHR cohort. Rare GPR37L1 coding variants were binned according to predicted pathogenicity and analyzed by sequence kernel association testing to reveal significant associations with disease diagnostic codes for epilepsy and migraine, among others. Since associations do not prove causality, rare GPR37L1 variants were functionally analyzed in SK-N-MC cells to evaluate potential signaling differences and pathogenicity. Notably, receptor variants exhibited varying abilities to reduce cAMP levels, activate mitogen-activated protein kinase (MAPK) signaling, and/or upregulate receptor expression in response to the agonist prosaptide (TX14(A)), as compared with the wild-type receptor. In addition to signaling changes, knock-out (KO) of GPR37L1 or expression of certain rare variants altered cellular cholesterol levels, which were also acutely regulated by administration of the agonist TX14(A) via activation of the MAPK pathway. Finally, to simulate the impact of rare nonsense variants found in the large patient cohort, a KO mouse line lacking Gpr37l1 was generated. Although KO animals did not recapitulate an acute migraine phenotype, the loss of this receptor produced sex-specific changes in anxiety-related disorders often seen in chronic migraineurs. Collectively, these observations define the existence of rare GPR37L1 variants associated with neuropsychiatric conditions in the human population and identify the signaling changes contributing to pathological processes.


Subject(s)
Migraine Disorders , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Migraine Disorders/genetics , Migraine Disorders/metabolism , Mice , Male , Female , Mice, Knockout , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Mice, Inbred C57BL , Genetic Variation/genetics
13.
J Integr Neurosci ; 23(4): 82, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38682225

ABSTRACT

BACKGROUND: Comorbid chronic neuropathic pain (NPP) and anxio-depressive disorders (ADD) have become a serious global public-health problem. The SLIT and NTRK-like 1 (SLITRK1) protein is important for synaptic remodeling and is highly expressed in the amygdala, an important brain region involved in various emotional behaviors. We examined whether SLITRK1 protein in the amygdala participates in NPP and comorbid ADD. METHODS: A chronic NPP mouse model was constructed by L5 spinal nerve ligation; changes in chronic pain and ADD-like behaviors were measured in behavioral tests. Changes in SLITRK1 protein and excitatory synaptic functional proteins in the amygdala were measured by immunofluorescence and Western blot. Adeno-associated virus was transfected into excitatory synaptic neurons in the amygdala to up-regulate the expression of SLITRK1. RESULTS: Chronic NPP-related ADD-like behavior was successfully produced in mice by L5 ligation. We found that chronic NPP and related ADD decreased amygdalar expression of SLITRK1 and proteins important for excitatory synaptic function, including Homer1, postsynaptic density protein 95 (PSD95), and synaptophysin. Virally-mediated SLITRK1 overexpression in the amygdala produced a significant easing of chronic NPP and ADD, and restored the expression levels of Homer1, PSD95, and synaptophysin. CONCLUSION: Our findings indicated that SLITRK1 in the amygdala plays an important role in chronic pain and related ADD, and may prove to be a potential therapeutic target for chronic NPP-ADD comorbidity.


Subject(s)
Amygdala , Behavior, Animal , Chronic Pain , Disks Large Homolog 4 Protein , Nerve Tissue Proteins , Neuralgia , Animals , Male , Mice , Amygdala/metabolism , Anxiety/metabolism , Anxiety/physiopathology , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Behavior, Animal/physiology , Chronic Pain/metabolism , Chronic Pain/physiopathology , Depression/metabolism , Depression/etiology , Depression/physiopathology , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Homer Scaffolding Proteins/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuralgia/metabolism , Synaptophysin/metabolism
14.
Physiol Behav ; 283: 114534, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38583548

ABSTRACT

The aim of this study was to investigate the effects of acupressure bladder meridian (ABM) on anxiety in rats. Chronic stress was induced rats to establish rat anxiety model. Shuttle experiment and open field experiments of were used to measure behaviors. The levels of cytokines in serum and hippocampus of rats were detected. Brain neurotransmitters (dopamine, 5- hydroxy tryptamine, norepinephrine) were detected by Enzyme linked immunosorbent assay (ELISA) kits. Immunohistochemistry and western blotting were used to detect MAPK and BDNF signal pathway in hippocampus of rats. ABM significantly improve behaviors, decreased cytokine levels in serum and hippocampus. ABM restored the changes of neurotransmitters and significantly decreased protein expressions of MAPK signal pathway and increased protein expressions of BDNF signal pathway in hippocampus of rats. The results shown that ABM significantly improved anxiety via inhibition of MAPK signal pathway and increased BDNF signal pathway.


Subject(s)
Brain-Derived Neurotrophic Factor , Hippocampus , Meridians , Signal Transduction , Animals , Male , Rats , Acupressure/methods , Anxiety Disorders/therapy , Anxiety Disorders/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cytokines/metabolism , Disease Models, Animal , Hippocampus/metabolism , MAP Kinase Signaling System/physiology , Rats, Sprague-Dawley , Signal Transduction/physiology , Urinary Bladder/metabolism
15.
Transl Psychiatry ; 14(1): 148, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38490997

ABSTRACT

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.


Subject(s)
Central Amygdaloid Nucleus , Humans , Animals , Female , Adolescent , Macaca mulatta , Temperament/physiology , Anxiety/metabolism , Anxiety Disorders/metabolism
16.
Neurobiol Dis ; 191: 106392, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38145853

ABSTRACT

Having experienced stress during sensitive periods of brain development strongly influences how individuals cope with later stress. Some are prone to develop anxiety or depression, while others appear resilient. The as-yet-unknown mechanisms underlying these differences may lie in how genes and environmental stress interact to shape the circuits that control emotions. Here, we investigated the role of the habenulo-interpeduncular system (HIPS), a critical node in reward circuits, in early stress-induced anxiety in mice. We found that habenular and IPN components characterized by the expression of Otx2 are synaptically connected and particularly sensitive to chronic stress (CS) during the peripubertal period. Stress-induced peripubertal activation of this HIPS subcircuit elicits both HIPS hypersensitivity to later stress and susceptibility to develop anxiety. We also show that HIPS silencing through conditional Otx2 knockout counteracts these effects of stress. Together, these results demonstrate that a genetic factor, Otx2, and stress interact during the peripubertal period to shape the stress sensitivity of the HIPS, which is shown to be a key modulator of susceptibility or resilience to develop anxiety.


Subject(s)
Habenula , Resilience, Psychological , Mice , Animals , Anxiety Disorders/metabolism , Emotions , Habenula/metabolism , Anxiety
17.
Cannabis Cannabinoid Res ; 8(6): 1045-1059, 2023 12.
Article in English | MEDLINE | ID: mdl-37862126

ABSTRACT

Background: Increasing evidence suggests that the endocannabinoid system (ECS) in the brain controls anxiety and may be a therapeutic target for the treatment of anxiety disorders. For example, both pharmacological and genetic disruption of cannabinoid receptor subtype-1 (CB1R) signaling in the central nervous system is associated with increased anxiety-like behaviors in rodents, while activating the system is anxiolytic. Sex is also a critical factor that controls the behavioral expression of anxiety; however, roles for the ECS in the gut in these processes and possible differences between sexes are largely unknown. Objective: In this study, we aimed to determine if CB1Rs in the intestinal epithelium exert control over anxiety-like behaviors in a sex-dependent manner. Methods: We subjected male and female mice with conditional deletion of CB1Rs in the intestinal epithelium (intCB1-/-) and controls (intCB1+/+) to the elevated plus maze (EPM), light/dark box, and open field test. Corticosterone (CORT) levels in plasma were measured at baseline and immediately after EPM exposure. Results: When compared with intCB1+/+ male mice, intCB1-/- male mice exhibited reduced levels of anxiety-like behaviors in the EPM and light/dark box. In contrast to male mice, no differences were found between female intCB1+/+ and intCB1-/- mice. Circulating CORT was higher in female versus male mice for both genotype groups at baseline and after EPM exposure; however, there was no effect of genotype on CORT levels. Conclusions: Collectively, these results indicate that genetic deletion of CB1Rs in the intestinal epithelium is associated with an anxiolytic phenotype in a sex-dependent manner.


Subject(s)
Anxiety Disorders , Anxiety , Receptor, Cannabinoid, CB1 , Animals , Female , Male , Mice , Anxiety/genetics , Anxiety/metabolism , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Corticosterone , Receptors, Cannabinoid/genetics , Receptors, Cannabinoid/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism
18.
Eur J Neurosci ; 57(12): 1966-1979, 2023 06.
Article in English | MEDLINE | ID: mdl-37165567

ABSTRACT

Anxiety disorders are the most prevalent co-morbidity factor associated with the core domains of autism spectrum disorders (ASD). Investigations on potential common neuronal mechanisms that may explain the co-occurrence of ASD and anxiety disorders are still poorly explored. One of the key questions that remained unsolved is the role of Shank3 protein in anxiety behaviours. Firstly, we characterize the developmental trajectories of locomotor, social behaviour and anxiety traits in a mouse model of ASD. We highlight that the anxiety phenotype is a late-onset emerging phenotype in mice with a Shank3Δe4-22 mutation. Consequently, we used an shRNA strategy to model Shank3 insufficiency in the bed nucleus of the stria terminalis (BNST), a brain region exerting a powerful control on anxiety level. We found that Shank3 downregulation in the anteromedial BNST (amBNST) induced anxiogenic effects and enhanced social avoidance after aversive social defeat. Associated with these behavioural defects, we showed alteration of glutamatergic synaptic functions in the amBNST induced by Shank3 insufficiency during adolescence. Our data strongly support the role of Shank3 in the maturation of amBNST, and its key role in anxiety control. Our results may further help to pave the road on a better understanding of the neuronal mechanisms underlying anxiety disorders implicated in ASDs.


Subject(s)
Septal Nuclei , Mice , Animals , Septal Nuclei/metabolism , Social Behavior , Anxiety/metabolism , Anxiety Disorders/metabolism , Phenotype , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
19.
Behav Brain Res ; 449: 114476, 2023 07 09.
Article in English | MEDLINE | ID: mdl-37148916

ABSTRACT

The prevalence of mental disorders such as depression and anxiety is high and often comorbid with other diseases. Chronic stress is a common risk factor for these disorders, but the mechanisms behind their development are not yet fully understood. Metabolomics has revealed a close association between purine and pyrimidine metabolism and depression and anxiety, with increased levels of serum xanthine observed in both humans and mice. Xanthine is known as purine metabolism, and this compound shows several biological activities, but the impact of xanthine on our brain function is still unclear. The hippocampus, which plays a crucial role in memory and learning, is also implicated in the pathophysiology of depression and anxiety. Here, we investigated the effects of xanthine intraperitoneal administration on spatial memory and anxiety-like behavior in mice. The findings indicated that xanthine administration induced a deficit of hippocampus-dependent spatial memory and a tendency to anxiety-like behavior in mice. RNA-seq analysis showed that xanthine administration upregulated hemoglobin (Hb) genes involved in oxygen transport in the hippocampus. The upregulated Hb genes occurred in the neuronal cells, and in vitro experiments revealed that both Hba-a1 derived from mice and HBA2 derived from humans were upregulated by xanthine treatment. These observations suggest that the xanthine-induced Hb in the hippocampus could be related to spatial memory deficit and anxiety. This study sheds light on the direct effects of xanthine on the brain and its potential role in the development of depression and anxiety symptoms caused by chronic stress.


Subject(s)
Anxiety Disorders , Anxiety , Humans , Mice , Animals , Xanthine/metabolism , Xanthine/pharmacology , Anxiety/metabolism , Anxiety Disorders/metabolism , Spatial Memory , Behavior, Animal/physiology , Hippocampus/metabolism
20.
Neuropsychopharmacology ; 48(10): 1532-1540, 2023 09.
Article in English | MEDLINE | ID: mdl-36949148

ABSTRACT

Differential expression of myelin-related genes and changes in myelin thickness have been demonstrated in mice after chronic psychosocial stress, a risk factor for anxiety disorders. To determine whether and how stress affects structural remodeling of nodes of Ranvier, another form of myelin plasticity, we developed a 3D reconstruction analysis of node morphology in C57BL/6NCrl and DBA/2NCrl mice. We identified strain-dependent effects of chronic social defeat stress on node morphology in the medial prefrontal cortex (mPFC) gray matter, including shortening of paranodes in C57BL/6NCrl stress-resilient and shortening of node gaps in DBA/2NCrl stress-susceptible mice compared to controls. Neuronal activity has been associated with changes in myelin thickness. To investigate whether neuronal activation is a mechanism influencing also node of Ranvier morphology, we used DREADDs to repeatedly activate the ventral hippocampus-to-mPFC pathway. We found reduced anxiety-like behavior and shortened paranodes specifically in stimulated, but not in the nearby non-stimulated axons. Altogether, our data demonstrate (1) nodal remodeling of the mPFC gray matter axons after chronic stress and (2) axon-specific regulation of paranodes in response to repeated neuronal activity in an anxiety-associated pathway. Nodal remodeling may thus contribute to aberrant circuit function associated with anxiety disorders.


Subject(s)
Anxiety Disorders , Anxiety , Mice , Animals , Mice, Inbred C57BL , Mice, Inbred DBA , Anxiety/metabolism , Anxiety Disorders/metabolism , Stress, Psychological/metabolism , Prefrontal Cortex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL