Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 807
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Sci Rep ; 13(1): 4970, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36973385

ABSTRACT

Microglia are immune brain cells implicated in stress-related mental illnesses including posttraumatic stress disorder (PTSD). Their role in the pathophysiology of PTSD, and on neurobiological systems that regulate stress, is not completely understood. We tested the hypothesis that microglia activation, in fronto-limbic brain regions involved in PTSD, would be elevated in participants with occupation-related PTSD. We also explored the relationship between cortisol and microglia activation. Twenty participants with PTSD and 23 healthy controls (HC) completed positron emission tomography (PET) scanning of the 18-kDa translocator protein (TSPO), a putative biomarker of microglia activation using the probe [18F]FEPPA, and blood samples for measurement of cortisol. [18F]FEPPA VT was non-significantly elevated (6.5-30%) in fronto-limbic regions in PTSD participants. [18F]FEPPA VT was significantly higher in PTSD participants reporting frequent cannabis use compared to PTSD non-users (44%, p = 0.047). Male participants with PTSD (21%, p = 0.094) and a history of early childhood trauma (33%, p = 0.116) had non-significantly higher [18F]FEPPA VT. Average fronto-limbic [18F]FEPPA VT was positively related to cortisol (r = 0.530, p = 0.028) in the PTSD group only. Although we did not find a significant abnormality in TSPO binding in PTSD, findings suggest microglial activation might have occurred in a subgroup who reported frequent cannabis use. The relationship between cortisol and TSPO binding suggests a potential link between hypothalamic-pituitary-adrenal-axis dysregulation and central immune response to trauma which warrants further study.


Subject(s)
Stress Disorders, Post-Traumatic , Child, Preschool , Humans , Male , Stress Disorders, Post-Traumatic/diagnostic imaging , Stress Disorders, Post-Traumatic/metabolism , Hydrocortisone/metabolism , Brain/diagnostic imaging , Brain/metabolism , Anxiety Disorders/metabolism , Positron-Emission Tomography/methods , Receptors, GABA/metabolism , Occupations
11.
Psychoneuroendocrinology ; 151: 106080, 2023 05.
Article in English | MEDLINE | ID: mdl-36931057

ABSTRACT

One negative emotional state from morphine protracted abstinence is anxiety which can drive craving and relapse risk in opioid addicts. Although the orexinergic system has been reported to be important in mediating emotion processing and addiction, the role of orexinergic system in anxiety from drug protracted abstinence remains elusive. In this study, by using behavioral test, western blot, electrophysiology and virus-mediated regulation of orexin receptor 1 (OX1R), we found that: (1) Intraperitoneal and intra-VTA administration of a selective OX1R antagonist SB334867 alleviated anxiety-like behaviors in open field test (OFT) but not in elevated plus maze test (EPM) in morphine protracted abstinent male mice. (2) OX1R expression in the VTA was upregulated by morphine withdrawal. (3) Virus-mediated knockdown of OX1R in the VTA prevented morphine abstinence-induced anxiety-like behaviors and virus-mediated overexpression of OX1R in the VTA was sufficient to produce anxiety-like behaviors in male mice. (4) The VTA neuronal activity was increased significantly induced by morphine protracted abstinence, which was mediated by OX1R. (5) OX1R was widely distributed in the neuronal soma and processes of dopaminergic and non-dopaminergic neurons in the VTA. The findings revealed that the OX1R mediates morphine abstinence-induced anxiety-like behaviors and the VTA plays a critical role in this effect.


Subject(s)
Anxiety , Morphine , Mice , Male , Animals , Morphine/pharmacology , Morphine/metabolism , Orexin Receptors/metabolism , Anxiety/metabolism , Anxiety Disorders/metabolism , Neurons/metabolism
12.
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
13.
Eur J Neurosci ; 57(6): 900-917, 2023 03.
Article in English | MEDLINE | ID: mdl-36725691

ABSTRACT

The bed nuclei of the stria terminalis (BST) is recognised as a pivotal integrative centre for monitoring emotional valence. It is implicated in the regulation of diverse affective states and motivated behaviours, and decades of research have firmly established its critical role in anxiety-related behavioural processes. Researchers have recently intricately dissected the BST's dynamic activities, its connection patterns and its functions with respect to specific cell types using multiple techniques such as optogenetics, in vivo calcium imaging and transgenic tools to unmask the complex circuitry mechanisms that underlie anxiety. In this review, we principally focus on studies of anxiety-involved neuromodulators within the BST and provide a comprehensive architecture of the anxiety network-highlighting the BST as a key hub in orchestrating anxiety-like behaviour. We posit that these promising efforts will contribute to the identification of an accurate roadmap for future treatment of anxiety disorders.


Subject(s)
Anxiety , Septal Nuclei , Animals , Humans , Anxiety/psychology , Anxiety Disorders/metabolism , Emotions , Animals, Genetically Modified , Septal Nuclei/metabolism
14.
Transl Psychiatry ; 13(1): 47, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36746925

ABSTRACT

Extracellular vesicles (EVs) are present in numerous peripheral bodily fluids and function in critical biological processes, including cell-to-cell communication. Most relevant to the present study, EVs contain microRNAs (miRNAs), and initial evidence from the field indicates that miRNAs detected in circulating EVs have been previously associated with mental health disorders. Here, we conducted an exploratory longitudinal and cross-sectional analysis of miRNA expression in serum EVs from adolescent participants. We analyzed data from a larger ongoing cohort study, evaluating 116 adolescent participants at two time points (wave 1 and wave 2) separated by three years. Two separate data analyses were employed: A cross-sectional analysis compared individuals diagnosed with Major Depressive Disorder (MDD), Anxiety disorders (ANX) and Attention deficit/Hyperactivity disorder (ADHD) with individuals without psychiatric diagnosis at each time point. A longitudinal analysis assessed changes in miRNA expression over time between four groups showing different diagnostic trajectories (persistent diagnosis, first incidence, remitted and typically developing/control). Total EVs were isolated, characterized by size distribution and membrane proteins, and miRNAs were isolated and sequenced. We then selected differentially expressed miRNAs for target prediction and pathway enrichment analysis. In the longitudinal analysis, we did not observe any statistically significant results. In the cross-sectional analysis: in the ADHD group, we observed an upregulation of miR-328-3p at wave 1 only; in the MDD group, we observed a downregulation of miR-4433b-5p, miR-584-5p, miR-625-3p, miR-432-5p and miR-409-3p at wave 2 only; and in the ANX group, we observed a downregulation of miR-432-5p, miR-151a-5p and miR-584-5p in ANX cases at wave 2 only. Our results identified previously observed and novel differentially expressed miRNAs and their relationship with three mental health disorders. These data are consistent with the notion that these miRNAs might regulate the expression of genes associated with these traits in genome-wide association studies. The findings support the promise of continued identification of miRNAs contained within peripheral EVs as biomarkers for mental health disorders.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Depressive Disorder, Major , Extracellular Vesicles , MicroRNAs , Humans , Adolescent , MicroRNAs/genetics , MicroRNAs/metabolism , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/metabolism , Cohort Studies , Cross-Sectional Studies , Depression , Genome-Wide Association Study , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism
15.
Int J Mol Sci ; 24(2)2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36674721

ABSTRACT

Klotho (KL) is a glycosyl hydrolase and aging-suppressor gene. Stress is a risk factor for depression and anxiety, which are highly comorbid with each other. The aim of this study is to determine whether KL is regulated by estrogen and plays an important role in sex differences in stress resilience. Our results showed that KL is regulated by estrogen in rat hippocampal neurons in vivo and in vitro and is essential for the estrogen-mediated increase in the number of presynaptic vesicular glutamate transporter 1 (Vglut1)-positive clusters on the dendrites of hippocampal neurons. The role of KL in sex differences in stress response was examined in rats using 3-week chronic unpredictable mild stress (CUMS). CUMS produced a deficit in spatial learning and memory, anhedonic-like behaviors, and anxiety-like behaviors in male but not female rats, which was accompanied by a reduction in KL protein levels in the hippocampus of male but not female rats. This demonstrated the resilience of female rats to CUMS. Interestingly, the knockdown of KL protein levels in the rat hippocampus of both sexes caused a decrease in stress resilience in both sexes, especially in female rats. These results suggest that the regulation of KL by estrogen plays an important role in estrogen-mediated synapse formation and that KL plays a critical role in the sex differences in cognitive deficit, anhedonic-like behaviors, and anxiety-like behaviors induced by chronic stress in rats, highlighting an important role of KL in sex differences in stress resilience.


Subject(s)
Depression , Sex Characteristics , Rats , Animals , Male , Female , Depression/metabolism , Anxiety , Anxiety Disorders/metabolism , Hippocampus/metabolism , Stress, Psychological/metabolism , Disease Models, Animal , Estrogens/metabolism
16.
Pharmacol Biochem Behav ; 223: 173513, 2023 02.
Article in English | MEDLINE | ID: mdl-36610590

ABSTRACT

Binge patterns of alcohol use, prevalent among adolescents, are associated with a higher probability of developing alcohol use disorders (AUD) and other psychiatric disorders, like anxiety and depression. Additionally, adverse life events strongly predict AUD and other psychiatric disorders. As such, the combined fields of stress and AUD have been well established, and animal models indicate that both binge-like alcohol exposure and stress exposure elevate anxiety-like behaviors. However, few have investigated the interaction of adolescent intermittent ethanol (AIE) and adult stressors. We hypothesized that AIE would increase vulnerability to restraint-induced stress (RS), manifested as increased anxiety-like behavior. After AIE exposure, in adulthood, animals were tested on forced swim (FST) and saccharin preference (SP) and then exposed to either RS (90 min/5 days) or home-cage control. Twenty-four hours after the last RS session, animals began testing on the elevated plus maze (EPM), and were re-tested on FST and SP. A separate group of animals were sacrificed in adulthood after AIE and RS, and brains were harvested for immunoblot analysis of dorsal and ventral hippocampus. Consistent with previous reports, AIE had no significant effect on closed arm time in the EPM (anxiety-like behavior). However, in male rats the interaction of AIE and adult RS increased time spent in the closed arms. No effect was observed among female animals. AIE and RS-specific alterations were found in glial and synaptic markers (GLT-1, FMRP and PSD-95) in male animals. These findings indicate AIE has sex-specific effects on both SP and the interaction of AIE and adult RS, which induces a propensity toward anxiety-like behavior in males. Also, AIE produces persistent hippocampal deficits that may interact with adult RS to cause increased anxiety-like behaviors. Understanding the mechanisms behind this AIE-induced increase in stress vulnerability may provide insight into treatment and prevention strategies for alcohol use disorders.


Subject(s)
Anxiety , Binge Drinking , Ethanol , Animals , Female , Male , Rats , Alcohol Drinking/adverse effects , Alcohol Drinking/metabolism , Alcohol Drinking/pathology , Alcohol Drinking/psychology , Alcoholism , Anxiety/etiology , Anxiety/metabolism , Anxiety/pathology , Anxiety/psychology , Anxiety Disorders/etiology , Anxiety Disorders/metabolism , Anxiety Disorders/pathology , Anxiety Disorders/psychology , Binge Drinking/complications , Binge Drinking/metabolism , Binge Drinking/pathology , Binge Drinking/psychology , Brain/drug effects , Brain/metabolism , Brain/pathology , Ethanol/adverse effects , Ethanol/pharmacology , Sex Factors , Stress, Psychological/complications , Stress, Psychological/metabolism , Stress, Psychological/pathology , Stress, Psychological/psychology
17.
Curr Neuropharmacol ; 21(9): 1864-1883, 2023.
Article in English | MEDLINE | ID: mdl-36237160

ABSTRACT

The Carioca rat lines originated from the selective bidirectional breeding of mates displaying extreme defense responses to contextual conditioned fear. After three generations, two distinct populations could be distinguished: the Carioca High- and Low-conditioned Freezing rats, CHF, and CLF, respectively. Later studies identified strong anxiety-like behaviors in the CHF line, while indications of impulsivity and hyperactivity were prominent in the CLF animals. The present review details the physiological and pharmacological-related findings obtained from these lines. The results discussed here point towards a dysfunctional fear circuitry in CHF rats, including alterations in key brain structures and the serotoninergic system. Moreover, data from these animals highlight important alterations in the stress-processing machinery and its associated systems, such as energy metabolism and antioxidative defense. Finally, evidence of an alteration in the dopaminergic pathway in CLF rats is also debated. Thus, accumulating data gathered over the years, place the Carioca lines as significant animal models for the study of psychiatric disorders, especially fear-related ones like anxiety.


Subject(s)
Anxiety , Fear , Rats , Animals , Anxiety/metabolism , Anxiety/psychology , Brain/metabolism , Anxiety Disorders/metabolism , Dopamine/metabolism
18.
Neuroscientist ; 29(4): 488-505, 2023 08.
Article in English | MEDLINE | ID: mdl-35086369

ABSTRACT

Dysfunction in the prefrontal cortex is commonly implicated in anxiety disorders, but the mechanisms remain unclear. Approach-avoidance conflict tasks have been extensively used in animal research to better understand how changes in neural activity within the prefrontal cortex contribute to avoidance behaviors, which are believed to play a major role in the maintenance of anxiety disorders. In this article, we first review studies utilizing in vivo electrophysiology to reveal the relationship between changes in neural activity and avoidance behavior in rodents. We then review recent studies that take advantage of optical and genetic techniques to test the unique contribution of specific prefrontal cortex circuits and cell types to the control of anxiety-related avoidance behaviors. This new body of work reveals that behavior during approach-avoidance conflict is dynamically modulated by individual cell types, distinct neural pathways, and specific oscillatory frequencies. The integration of these different pathways, particularly as mediated by interactions between excitatory and inhibitory neurons, represents an exciting opportunity for the future of understanding anxiety.


Subject(s)
Anxiety Disorders , Anxiety , Animals , Anxiety Disorders/metabolism , Prefrontal Cortex/physiology , Avoidance Learning/physiology , Neural Pathways
19.
Front Neuroendocrinol ; 67: 101030, 2022 10.
Article in English | MEDLINE | ID: mdl-35995079

ABSTRACT

Exposure therapy is a central component of the first-line treatment for anxiety disorders, a common mental health condition that is twice as prevalent in women relative to men. A key underlying mechanism of exposure therapy is fear extinction, which is an active learning process supported by a neural circuitry that is highly regulated by ovarian hormones. This review synthesises research examining the impact of hormonal contraceptives on laboratory fear extinction tasks in female rats and women, and on exposure therapy in women with anxiety disorders. The evidence indicates that hormonal contraceptives have a detrimental impact on fear extinction and exposure therapy that is consistent across species, and from laboratory to clinical settings. Candidate pathways by which hormonal contraceptives impede fear extinction and exposure therapy include suppression of endogenous ovarian hormones and glucocorticoids, and downregulation of signalling pathways that support extinction learning. Key areas of focus for future research are discussed.


Subject(s)
Extinction, Psychological , Fear , Female , Animals , Rats , Extinction, Psychological/physiology , Fear/physiology , Contraceptive Agents , Anxiety/drug therapy , Anxiety/metabolism , Anxiety Disorders/metabolism , Hormones
20.
Neurosci Lett ; 788: 136851, 2022 09 25.
Article in English | MEDLINE | ID: mdl-36007708

ABSTRACT

Post-traumatic stress disorder (PTSD) has become epidemic following severely stressful incidents. Previous studies have shown that brain-derived neurotrophic factor (BDNF) has anxiolytic effects on various anxiety or depression disorders including PTSD. However, the detailed mechanisms of BDNF for treating PTSD were rarely investigated. In the current study, single-prolonged stress (SPS) was used as an animal model recapitulating specific aspects for a PTSD-like phenotype. The effects of BDNF on SPS-induced anxiety-like behaviors were investigated. We showed that the levels of BDNF in the cerebro-spinal fluid (CSF) were significantly reduced after the rats experienced SPS. The SPS-induced reductions of percentages of time spent in the central area to total time in the open field test, were dose-dependently mitigated after BDNF intracerebroventricular (i.c.v.) injections. BDNF i.c.v. administration also dose-dependently increased the preference of the light box in the light-dark box test. Both expressions of tyrosine kinase receptor B (TrkB) protein and mRNA in the prefrontal cortex (PFC) and amygdala were significantly increased after SPS challenges. BDNF i.c.v. administration attenuated these compensatory increases of TrkB. At last, the anxiolytic effects of BDNF on SPS model were also observed by using other two injection methods. These results inspired us to study that different administrations of BDNF were used in patients with PTSD in the future, in-depthly.


Subject(s)
Anti-Anxiety Agents , Stress Disorders, Post-Traumatic , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Anxiety Disorders/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Hippocampus/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...