Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 9.422
1.
Dev Psychobiol ; 66(5): e22512, 2024 Jul.
Article En | MEDLINE | ID: mdl-38837366

The current study examined associations between parental adversities as experienced in adolescence and hair cortisol concentration (HCC) 26 years later (n = 47). Specifically, bivariate correlations and linear regressions were used to examine harsh parenting as well as parental economic pressure, emotional distress, and body mass index (BMI) when their adolescent was between 15 and 16 years old (parent average age 43). HCC was measured when the adolescent was an adult (average 42 years old), at a similar age to when their parent(s) first participated in the study. We also assessed their economic pressure, emotional distress, obesity, and perceived stress in adulthood. For results across generations, parental economic pressure experienced during adolescence was significantly related to HCC when these adolescents were adults. None of the adult economic pressure, emotional distress, BMI, and perceived stress variables were associated with their HCC. Interestingly, there were significant associations among adult perceived stress, economic pressure, emotional distress, and obesity. Thus, the association between parental economic pressure and adult HCC is independent of adult adversities. Results highlight early economic adversity as a possible childhood stressor that has implications throughout the life course.


Adverse Childhood Experiences , Hair , Hydrocortisone , Humans , Hair/chemistry , Female , Male , Hydrocortisone/metabolism , Hydrocortisone/analysis , Adult , Adolescent , Stress, Psychological/metabolism , Parenting , Body Mass Index , Psychological Distress , Obesity/metabolism
2.
Dev Psychobiol ; 66(5): e22511, 2024 Jul.
Article En | MEDLINE | ID: mdl-38837722

Patients diagnosed with posttraumatic stress disorder (PTSD) present with a spectrum of debilitating anxiety symptoms resulting from exposure to trauma. Women are twice as likely to be diagnosed with anxiety and PTSD compared to men; however, the reason for this vulnerability remains unknown. We conducted four experiments where we first demonstrated a female vulnerability to stress-enhanced fear learning (SEFL) with a moderate, acute early life stress (aELS) exposure (4 footshocks in a single session), compared to a more intense aELS exposure (15 footshocks in a single session) where males and females demonstrated comparable SEFL. Next, we demonstrated that this female vulnerability does not result from differences in footshock reactivity or contextual fear conditioning during the aELS exposure. Finally, using gonadectomy or sham surgeries in adult male and female rats, we showed that circulating levels of gonadal steroid hormones at the time of adult fear conditioning do not explain the female vulnerability to SEFL. Additional research is needed to determine whether this vulnerability can be explained by organizational effects of gonadal steroid hormones or differences in sex chromosome gene expression. Doing so is critical for a better understanding of increased female vulnerability to certain psychiatric diseases.


Fear , Sex Characteristics , Stress, Psychological , Animals , Fear/physiology , Male , Female , Rats , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Behavior, Animal/physiology , Conditioning, Classical/physiology , Rats, Sprague-Dawley , Gonadal Steroid Hormones/metabolism , Learning/physiology
3.
Stress ; 27(1): 2345906, 2024 Jan.
Article En | MEDLINE | ID: mdl-38841737

Mindfulness-based interventions have become a popular means to reduce stress. However, the specific mechanisms driving observed stress reduction remain understudied. The Monitor and Acceptance Theory suggests that the cultivation of monitoring and acceptance skills are necessary moderators of practice-induced stress reduction. In the context of the ReSource Project, a large healthy adult sample underwent three 3-month mental training modules targeting either attentional (Presence module), socio-affective (Affect module) or socio-cognitive skills (Perspective module). In the current study, the development of a range of inter-individual differences in mindfulness-, interoception- and compassion-related traits - which mapped to either monitoring or acceptance categories - was tracked. The relationship of these training-induced changes with cortisol stress reactivity after the three distinct 3-month training modules was explored. We found that stress sensitivity was particularly modulated by a differential adaptivity of one cultivated attentional capacity - Attention regulation - which predicted higher cortisol reactivity after mere attention training (Presence) but was associated with lower stress-induced cortisol release after additional socio-affective and socio-cognitive practice (Affect and Perspective). However, this effect did not survive multiple comparisons correction, and analyses were limited by the sample size available. We conclude that our study provides preliminary support of the Monitor and Acceptance Theory, lending weight to the advantage of primary attentional increases in order to fully harness the beneficial effects of socio-affective training, ultimately leading to stress reduction. Although training-induced increases in acceptance were not directly shown to contribute to lowering cortisol stress reactivity, the data suggest an additional benefit of socio-affective and socio-cognitive training that is not directly captured within the current analyses. Our study corroborates the importance of going beyond the training of attention monitoring to foster stress resilience, and highlights that mental training relies on the co-development of several interacting processes to successfully attenuate stress. Further exploring the overarching concept of acceptance in future research may prove beneficial to the theoretical framework of MAT, and in understanding the processes by which stress reduction occurs.


Attention , Hydrocortisone , Mindfulness , Stress, Psychological , Humans , Hydrocortisone/metabolism , Male , Stress, Psychological/metabolism , Female , Attention/physiology , Adult , Young Adult , Saliva/metabolism , Saliva/chemistry , Empathy/physiology , Interoception/physiology
4.
Transl Psychiatry ; 14(1): 239, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38834575

Prior research has identified differential protein expression levels of linker histone H1x within the ventral hippocampus (vHipp) of stress-susceptible versus stress-resilient mice. These mice are behaviorally classified based on their divergent responses to chronic social stress. Here, we sought to determine whether elevated vHipp H1x protein levels directly contribute to these diverging behavioral adaptations to stress. First, we demonstrated that stress-susceptible mice uniquely express elevated vHipp H1x protein levels following chronic stress. Given that linker histones coordinate heterochromatin compaction, we hypothesize that elevated levels of H1x in the vHipp may impede pro-resilience transcriptional adaptations and prevent development of the resilient phenotype following social stress. To test this, 8-10-week-old male C57BL/6 J mice were randomly assigned to groups undergoing 10 days of chronic social defeat stress (CSDS) or single housing, respectively. Following CSDS, mice were classified as susceptible versus resilient based on their social interaction behaviors. We synthesized a viral overexpression (OE) vector for H1x and transduced all stressed and single housed mice with either H1x or control GFP within vHipp. Following viral delivery, we conducted social, anxiety-like, and memory-reliant behavior tests on distinct cohorts of mice. We found no behavioral adaptations following H1x OE compared to GFP controls in susceptible, resilient, or single housed mice. In sum, although we confirm elevated vHipp protein levels of H1x associate with susceptibility to social stress, we observe no significant behavioral consequence of H1x OE. Thus, we conclude elevated levels of H1x are associated with, but are not singularly sufficient to drive development of behavioral adaptations to stress.


Behavior, Animal , Hippocampus , Histones , Mice, Inbred C57BL , Stress, Psychological , Animals , Male , Hippocampus/metabolism , Mice , Stress, Psychological/metabolism , Histones/metabolism , Behavior, Animal/physiology , Adaptation, Psychological/physiology , Resilience, Psychological , Social Defeat , Anxiety/metabolism
5.
Sci Adv ; 10(19): eadk7636, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728397

Corticotropin releasing factor (CRF) network in the oval nucleus of bed nuclei of the stria terminalis (ovBNST) is generally indicated in stress, but its role in female-biased susceptibility to anxiety is unknown. Here, we established a female-biased stress paradigm. We found that the CRF release in ovBNST during stress showed female-biased pattern, and ovBNST CRF neurons were more prone to be hyperexcited in female mice during stress in both in vitro and in vivo studies. Moreover, optogenetic modulation to exchange the activation pattern of ovBNST CRF neurons during stress between female and male mice could reverse their susceptibility to anxiety. Last, CRF receptor type 1 (CRFR1) mediated the CRF-induced excitation of ovBNST CRF neurons and showed female-biased expression. Specific knockdown of the CRFR1 level in ovBNST CRF neurons in female or overexpression that in male could reverse their susceptibility to anxiety. Therefore, we identify that CRFR1-mediated hyperexcitation of ovBNST CRF neurons in female mice encode the female-biased susceptibility to anxiety.


Anxiety , Corticotropin-Releasing Hormone , Neurons , Receptors, Corticotropin-Releasing Hormone , Septal Nuclei , Animals , Female , Anxiety/metabolism , Male , Neurons/metabolism , Corticotropin-Releasing Hormone/metabolism , Septal Nuclei/metabolism , Mice , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Avoidance Learning/physiology , Stress, Psychological/metabolism , Behavior, Animal
6.
Dev Psychobiol ; 66(5): e22494, 2024 Jul.
Article En | MEDLINE | ID: mdl-38698641

Though considerable work supports the Dimensional Model of Adversity and Psychopathology, prior research has not tested whether the dimensions-threat (e.g., abuse) and deprivation (e.g., neglect)-are uniquely related to salivary trait indicators of hypothalamic pituitary adrenal (HPA) axis activity. We examined the unique and interactive effects of threat and deprivation on latent trait cortisol (LTC)-and whether these effects were modified by co-occurring adversities. Emerging adults (n = 90; Mage = 19.36 years; 99.88% cisgender women) provided salivary cortisol samples four times a day (waking, 30 min and 45 min postwaking, bedtime) over three 3-day measurement waves over 13 weeks. Contextual life stress interviews assessed early adversity. Though the effects varied according to the conceptualization of early adversity, overall, threat-but not deprivation, nor other co-occurring adversities-was uniquely associated with the across-wave LTC. Specifically, the incidence and frequency of threat were each negatively related to the across-wave LTC. Threat severity was also associated with the across-wave LTC, but only among those with no deprivation. Finally, the effects of threat were modified by other co-occurring adversities. Findings suggest that threat has unique implications for individual differences in HPA axis activity among emerging adults, and that co-occurring adversities modify such effects.


Hydrocortisone , Hypothalamo-Hypophyseal System , Saliva , Humans , Female , Male , Hydrocortisone/metabolism , Young Adult , Adult , Saliva/metabolism , Saliva/chemistry , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Adolescent , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Adverse Childhood Experiences , Psychosocial Deprivation
7.
Cell Mol Life Sci ; 81(1): 205, 2024 May 04.
Article En | MEDLINE | ID: mdl-38703204

BACKGROUND: Exposure to chronic psychological stress (CPS) is a risk factor for thrombotic cardiocerebrovascular diseases (CCVDs). The expression and activity of the cysteine cathepsin K (CTSK) are upregulated in stressed cardiovascular tissues, and we investigated whether CTSK is involved in chronic stress-related thrombosis, focusing on stress serum-induced endothelial apoptosis. METHODS AND RESULTS: Eight-week-old wild-type male mice (CTSK+/+) randomly divided to non-stress and 3-week restraint stress groups received a left carotid artery iron chloride3 (FeCl3)-induced thrombosis injury for biological and morphological evaluations at specific timepoints. On day 21 post-stress/injury, the stress had enhanced the arterial thrombi weights and lengths, in addition to harmful alterations of plasma ADAMTS13, von Willebrand factor, and plasminogen activation inhibitor-1, plus injured-artery endothelial loss and CTSK protein/mRNA expression. The stressed CTSK+/+ mice had increased levels of injured arterial cleaved Notch1, Hes1, cleaved caspase8, matrix metalloproteinase-9/-2, angiotensin type 1 receptor, galactin3, p16IN4A, p22phox, gp91phox, intracellular adhesion molecule-1, TNF-α, MCP-1, and TLR-4 proteins and/or genes. Pharmacological and genetic inhibitions of CTSK ameliorated the stress-induced thrombus formation and the observed molecular and morphological changes. In cultured HUVECs, CTSK overexpression and silencing respectively increased and mitigated stressed-serum- and H2O2-induced apoptosis associated with apoptosis-related protein changes. Recombinant human CTSK degraded γ-secretase substrate in a dose-dependent manor and activated Notch1 and Hes1 expression upregulation. CONCLUSIONS: CTSK appeared to contribute to stress-related thrombosis in mice subjected to FeCl3 stress, possibly via the modulation of vascular inflammation, oxidative production and apoptosis, suggesting that CTSK could be an effective therapeutic target for CPS-related thrombotic events in patients with CCVDs.


Apoptosis , Cathepsin K , Chlorides , Disease Models, Animal , Ferric Compounds , Thrombosis , Animals , Humans , Male , Mice , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/genetics , Cathepsin K/metabolism , Cathepsin K/genetics , Chlorides/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/genetics , Stress, Psychological/complications , Stress, Psychological/metabolism , Thrombosis/metabolism , Thrombosis/pathology , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics
8.
Front Public Health ; 12: 1289689, 2024.
Article En | MEDLINE | ID: mdl-38813430

Introduction: Increasing evidence supports chronic psychological stress as a risk factor for the development of type 2 diabetes. Much less is known, however, about the role of chronic stress in established diabetes. Methods: The aim of the current study was to comprehensively assess chronic stress in a sample of 73 patients with type 2 diabetes and 48 non-diabetic control participants, and to investigate associations with indicators of glycemic control (HbA1c), insulin resistance (HOMA-IR), ß-cell functioning (C-peptide), illness duration, and the presence of microvascular complications. Chronic stress was measured using questionnaires [the Perceived Stress Scale (PSS), the Screening Scale of the Trier Inventory of Chronic Stress (SSCS), the Perceived Health Questionnaire (PHQ) as well as the Questionnaire on Stress in Patients with Diabetes-Revised (QSD-R)]; hair cortisol was used as a biological indicator. Results: We found that patients with type 2 diabetes had higher levels of hair cortisol in comparison to the control group (F(1,112) = 5.3; p = 0.023). Within the diabetic group, higher hair cortisol was associated with a longer duration of the illness (r = 0.25, p = 0.04). General perceived stress did not show significant associations with metabolic outcomes in type 2 diabetes patients. In contrast, higher diabetes-related distress, as measured with the QSD-R, was associated with lower glycemic control (r = 0.28, p = 0.02), higher insulin resistance (r = 0.26, p = 0.03) and a longer duration of the illness (r = 0.30, p = 0.01). Discussion: Our results corroborate the importance of chronic psychological stress in type 2 diabetes. It appears, however, that once type 2 diabetes has developed, diabetes-specific distress gains in importance over general subjective stress. On a biological level, increased cortisol production could be linked to the course of the illness.


Diabetes Mellitus, Type 2 , Hair , Hydrocortisone , Stress, Psychological , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/psychology , Diabetes Mellitus, Type 2/complications , Male , Female , Hydrocortisone/metabolism , Hydrocortisone/analysis , Middle Aged , Stress, Psychological/metabolism , Cross-Sectional Studies , Hair/metabolism , Hair/chemistry , Aged , Surveys and Questionnaires , Insulin Resistance , Adult , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , Risk Factors
9.
Mol Brain ; 17(1): 30, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802853

The Hypothalmic-Pituitary-Adrenal axis also known as the HPA axis is central to stress response. It also acts as the relay center between the body and the brain. We analysed hypothalamic proteome from mice subjected to chronic social defeat paradigm using iTRAQ based quantitative proteomics to identify changes associated with stress response. We identified greater than 2000 proteins after processing our samples analysed through Q-Exactive (Thermo) and Orbitrap Velos (Thermo) at 5% FDR. Analysis of data procured from the runs showed that the proteins whose levels were affected belonged primarily to mitochondrial and metabolic processes, translation, complement pathway among others. We also found increased levels of fibrinogen, myelin basic protein (MBP) and neurofilaments (NEFL, NEFM, NEFH) in the hypothalamus from socially defeated mice. Interestingly, research indicates that these proteins are upregulated in blood and CSF of subjects exposed to trauma and stress. Since hypothalamus secreted proteins can be found in blood and CSF, their utility as biomarkers in depression holds an impressive probability and should be validated in clinical samples.


Hypothalamus , Mice, Inbred C57BL , Social Defeat , Stress, Psychological , Animals , Hypothalamus/metabolism , Stress, Psychological/metabolism , Stress, Psychological/blood , Male , Proteomics/methods , Mice , Proteome/metabolism
10.
Bull Exp Biol Med ; 176(5): 612-616, 2024 Mar.
Article En | MEDLINE | ID: mdl-38730106

We experimentally demonstrated that chronic social stress during the development of a depression-like state enhances lung metastasis and modifies the expression of many carcinogenesis- and apoptosis-related genes in the hypothalamus of mice, including genes involved in lung cancer pathogenesis in humans. Analysis of the expression of genes encoding the major clinical markers of lung cancer in the hypothalamus of mice with depression-like behavior revealed increased expression of the Eno2 gene encoding neuron-specific enolase, a blood marker of lung cancer progression in humans. It was shown that the expression of this gene in the hypothalamus correlated with the expression of many carcinogenesis- and apoptosis-related genes. The discovered phenomenon may have a fundamental significance and requires further studies.


Apoptosis , Carcinogenesis , Depression , Hypothalamus , Lung Neoplasms , Phosphopyruvate Hydratase , Animals , Mice , Hypothalamus/metabolism , Hypothalamus/pathology , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Apoptosis/genetics , Depression/genetics , Depression/metabolism , Depression/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Carcinogenesis/genetics , Male , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism
11.
Stress ; 27(1): 2357330, 2024 Jan.
Article En | MEDLINE | ID: mdl-38775373

Why individuals suffer negative consequences following stress is a complex phenomenon that is dictated by individual factors, the timing of stress within the lifespan, and when in the lifespan the consequences are measured. Women who undergo adverse childhood experiences are at risk for lasting biological consequences, including affective and stress dysregulation. We have shown that pubertal adversity is associated with a blunted hypothalamic-pituitary-adrenal axis glucocorticoid response in peripartum humans and mice. In mice, our prior examination of the paraventricular nucleus (PVN) of the hypothalamus showed that pubertal stress led to an upregulation of baseline mRNA expression of six immediate early genes (IEGs) in the PVN of adult, pregnant mice. Separately, we showed that the pregnancy-associated hormone allopregnanolone is necessary and sufficient to produce the blunted stress response phenotype in pubertally stressed mice. In the current study, we further examined a potential mechanistic role for the IEGs in the PVN. We found that in pubertally stressed adult female, but not male, mice, intra-PVN allopregnanolone was sufficient to recapitulate the baseline IEG mRNA expression profile previously observed in pubertally stressed, pregnant mice. We also examined baseline IEG mRNA expression during adolescence, where we found that IEGs have developmental trajectories that showed sex-specific disruption by pubertal stress. Altogether, these data establish that IEGs may act as a key molecular switch involved in increased vulnerability to negative outcomes in adult, pubertally stressed animals. How the factors that produce vulnerability combine throughout the lifespan is key to our understanding of the etiology of stress-related disorders.


Paraventricular Hypothalamic Nucleus , Stress, Psychological , Transcriptome , Animals , Female , Male , Mice , Stress, Psychological/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Pregnanolone , Hypothalamus/metabolism , Hypothalamus/drug effects , Pregnancy , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/drug effects , Sexual Maturation , Genes, Immediate-Early
12.
Behav Brain Res ; 468: 115039, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38718877

Chronic unpredictable mild stress (CUMS) method has been introduced as a rodent model of depression. On the other hand, olanzapine, as an antipsychotic, can induce antidepressant and antipsychotic effects. Also, olanzapine may improve cognitive functions. Both CUMS and olanzapine can also affect the expression level of brain-derived neurotrophic factor (BDNF) and synaptophysin, the molecular factors involved in synaptic function, and learning and memory. In this study, we investigated the effect of olanzapine on locomotor activity (using open field test), pain threshold (using hot plate), depressive-like behavior (using forced swim test), spatial learning and memory (using Morris water maze), and BDNF and synaptophysin hippocampal expression (using real-time PCR) in both male and female CUMS rats. CUMS was performed for three consecutive weeks. Olanzapine was also injected intraperitoneally at the dose of 5 mg/kg. Our data showed that olanzapine can reverse the effects of CUMS on behavioral functions and BDNF and synaptophysin expression levels in the hippocampus of both males and females. It was also shown that olanzapine effects on spatial memory, pain perception, and BDNF and synaptophysin level were stronger in females than males. In conclusion, we suggested that the therapeutic effects of olanzapine in CUMS rats may be closely related to the function of BDNF and synaptophysin. Also, the therapeutic effects of olanzapine may be stronger in females. Therefore, and for the first time, we showed that there may be a sex difference in the effects of olanzapine on behavioral and molecular changes following CUMS.


Brain-Derived Neurotrophic Factor , Depression , Disease Models, Animal , Hippocampus , Olanzapine , Pain Perception , Spatial Memory , Stress, Psychological , Synaptophysin , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Male , Synaptophysin/metabolism , Female , Olanzapine/pharmacology , Stress, Psychological/metabolism , Stress, Psychological/drug therapy , Rats , Depression/drug therapy , Depression/metabolism , Spatial Memory/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Pain Perception/drug effects , Pain Perception/physiology , Behavior, Animal/drug effects , Memory Disorders/drug therapy , Memory Disorders/metabolism , Antipsychotic Agents/pharmacology , Rats, Sprague-Dawley
13.
Behav Brain Res ; 468: 115028, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38723677

Early life stress (ELS) increases the risk of depression later in life. Programmed cell death factor 4 (PDCD4), an apoptosis-related molecule, extensively participates in tumorigenesis and inflammatory diseases. However, its involvement in a person's susceptibility to ELS-related depression is unknown. To examine the effects and underlying mechanisms of PDCD4 on ELS vulnerability, we used a "two-hit" stress mouse model: an intraperitoneal injection of lipopolysaccharide (LPS) into neonatal mice was performed on postnatal days 7-9 (P7-P9) and inescapable foot shock (IFS) administration in adolescent was used as a later-life challenge. Our study shows that compared with mice that were only exposed to the LPS or IFS, the "two-hit" stress mice developed more severe depression/anxiety-like behaviors and social disability. We detected the levels of PDCD4 in the hippocampus of adolescent mice and found that they were significantly increased in "two-hit" stress mice. The results of immunohistochemical staining and Sholl analysis showed that the number of microglia in the hippocampus of "two-hit" stress mice significantly increased, with morphological changes, shortened branches, and decreased numbers. However, knocking down PDCD4 can prevent the number and morphological changes of microglia induced by ELS. In addition, we confirmed through the Golgi staining and immunohistochemical staining results that knocking down PDCD4 can ameliorate ELS-induced synaptic plasticity damage. Mechanically, the knockdown of PDCD4 exerts neuroprotective effects, possibly via the mediation of BDNF/AKT/CREB signaling. Combined, these results suggest that PDCD4 may play an important role in the ELS-induced susceptibility to depression and, thus, may become a therapeutic target for depressive disorders.


Apoptosis Regulatory Proteins , Depression , Hippocampus , Mice, Inbred C57BL , Neuronal Plasticity , RNA-Binding Proteins , Stress, Psychological , Animals , Hippocampus/metabolism , Neuronal Plasticity/physiology , Stress, Psychological/metabolism , Mice , Depression/metabolism , Depression/physiopathology , Apoptosis Regulatory Proteins/metabolism , RNA-Binding Proteins/metabolism , Male , Disease Models, Animal , Microglia/metabolism , Lipopolysaccharides/pharmacology , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Disease Susceptibility , Animals, Newborn
14.
Biomolecules ; 14(5)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38785934

Adverse experiences (e.g., acute stress) and alcohol misuse can both impair skeletal muscle homeostasis, resulting in reduced protein synthesis and greater protein breakdown. Exposure to acute stress is a significant risk factor for engaging in alcohol misuse. However, little is known about how these factors together might further affect skeletal muscle health. To that end, this study investigated the effects of acute stress exposure followed by a period of binge-patterned alcohol drinking on signaling factors along mouse skeletal muscle protein synthesis (MPS) and degradation (MPD) pathways. Young adult male C57BL/6J mice participated in the Drinking in the Dark paradigm, where they received 2-4 h of access to 20% ethanol (alcohol group) or water (control group) for four days to establish baseline drinking levels. Three days later, half of the mice in each group were either exposed to a single episode of uncontrollable tail shocks (acute stress) or remained undisturbed in their home cages (no stress). Three days after stress exposure, mice received 4 h of access to 20% ethanol (alcohol) to model binge-patterned alcohol drinking or water for ten consecutive days. Immediately following the final episode of alcohol access, mouse gastrocnemius muscle was extracted to measure changes in relative protein levels along the Akt-mTOR MPS, as well as the ubiquitin-proteasome pathway (UPP) and autophagy MPD pathways via Western blotting. A single exposure to acute stress impaired Akt singling and reduced rates of MPS, independent of alcohol access. This observation was concurrent with a potent increase in heat shock protein seventy expression in the muscle of stressed mice. Alcohol drinking did not exacerbate stress-induced alterations in the MPS and MPD signaling pathways. Instead, changes in the MPS and MPD signaling factors due to alcohol access were primarily observed in non-stressed mice. Taken together, these data suggest that exposure to a stressor of sufficient intensity may cause prolonged disruptions to signaling factors that impact skeletal muscle health and function beyond what could be further induced by periods of alcohol misuse.


Binge Drinking , Mice, Inbred C57BL , Muscle Proteins , Muscle, Skeletal , Proteolysis , Animals , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Mice , Muscle Proteins/metabolism , Muscle Proteins/biosynthesis , Binge Drinking/metabolism , Proteolysis/drug effects , Signal Transduction/drug effects , Protein Biosynthesis/drug effects , Ethanol , Stress, Psychological/metabolism , TOR Serine-Threonine Kinases/metabolism , Alcohol Drinking/metabolism
15.
Dev Psychobiol ; 66(5): e22502, 2024 Jul.
Article En | MEDLINE | ID: mdl-38807271

Environmental influences before and during pregnancy significantly impact offspring development. This study investigates open research questions regarding the associations between maternal early life stress (ELS), prenatal psychosocial stress, prenatal hair cortisol (HC), and birth outcomes in Argentinian women. Data on ELS, prenatal life events, HC (two samples representing first and second half of pregnancy), and birth outcomes were collected from middle-class Argentinian women (N = 69) upon delivery. Linear mixed models indicated that HC increased from the first half to the second half of pregnancy with considerable variability in the starting values and slopes between individuals. Mothers who experienced more ELS, were taller, or more educated, tended to show lower increases in HC. Older age was positively related to HC increases. Our data did not suggest an interaction between ELS and prenatal life events in relation to HC. We found that the change in HC was most likely negatively associated with birth weight. Our data are most compatible with either a weak or the absence of an association between ELS or prenatal life events and absolute values of HC. Mothers with stronger increases in hair cortisol tended to have newborns with slightly lower birth weight. Hence, ELS and birthweight may either have been related to changes in cortisol exposure during pregnancy or to factors that influence accumulation or retention of cortisol in hair.


Birth Weight , Hair , Hydrocortisone , Prenatal Exposure Delayed Effects , Stress, Psychological , Humans , Female , Pregnancy , Hydrocortisone/metabolism , Hydrocortisone/analysis , Stress, Psychological/metabolism , Adult , Hair/chemistry , Argentina , Birth Weight/physiology , Infant, Newborn , Pregnancy Outcome , Young Adult , Mothers
16.
Int J Mol Sci ; 25(10)2024 May 07.
Article En | MEDLINE | ID: mdl-38791125

The brain is the central organ of adaptation to stress because it perceives and determines threats that induce behavioral, physiological, and molecular responses. In humans, chronic stress manifests as an enduring consistent feeling of pressure and being overwhelmed for an extended duration. This can result in a persistent proinflammatory response in the peripheral and central nervous system (CNS), resulting in cellular, physiological, and behavioral effects. Compounding stressors may increase the risk of chronic-stress-induced inflammation, which can yield serious health consequences, including mental health disorders. This review summarizes the current knowledge surrounding the neuroinflammatory response in rodent models of chronic stress-a relationship that is continually being defined. Many studies investigating the effects of chronic stress on neuroinflammation in rodent models have identified significant changes in inflammatory modulators, including nuclear factor-κB (NF-κB) and toll-like receptors (TLRs), and cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1ß, and IL-6. This suggests that these are key inflammatory factors in the chronic stress response, which may contribute to the establishment of anxiety and depression-like symptoms. The behavioral and neurological effects of modulating inflammatory factors through gene knockdown (KD) and knockout (KO), and conventional and alternative medicine approaches, are discussed.


Disease Models, Animal , Neuroinflammatory Diseases , Stress, Psychological , Animals , Humans , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Stress, Psychological/metabolism , Rodentia , Chronic Disease , Cytokines/metabolism , NF-kappa B/metabolism , Inflammation/metabolism
17.
Pharmacol Res ; 204: 107214, 2024 Jun.
Article En | MEDLINE | ID: mdl-38763328

Studies have shown that the microbiota-gut-brain axis is highly correlated with the pathogenesis of depression in humans. However, whether independent oral microbiome that do not depend on gut microbes could affect the progression of depression in human beings remains unclear, neither does the presence and underlying mechanisms of the microbiota-oral-brain axis in the development of the condition. Hence this study that encompasses clinical and animal experiments aims at investigating the correlation between oral microbiota and the onset of depression via mediating the microbiota-oral-brain axis. We compared the oral microbial compositions and metabolomes of 87 patients with depressive symptoms versus 70 healthy controls. We found that the oral microbial and metabolic signatures were significantly different between the two groups. Significantly, germ-free (GF) mice transplanted with saliva from mice exposing to chronic restraint stress (CRS) displayed depression-like behavior and oral microbial dysbiosis. This was characterized by a significant differential abundance of bacterial species, including the enrichment of Pseudomonas, Pasteurellaceae, and Muribacter, as well as the depletion of Streptococcus. Metabolomic analysis showed the alternation of metabolites in the plasma of CRS-exposed GF mice, especially Eicosapentaenoic Acid. Furthermore, oral and gut barrier dysfunction caused by CRS-induced oral microbiota dysbiosis may be associated with increased blood-brain barrier permeability. Pseudomonas aeruginosa supplementation exacerbated depression-like behavior, while Eicosapentaenoic Acid treatment conferred protection against depression-like states in mice. These results suggest that oral microbiome and metabolic function dysbiosis may be relevant to the pathogenesis and pathophysiology of depression. The proposed microbiota-oral-brain axis provides a new way and targets for us to study the pathogenesis of depression.


Depression , Dysbiosis , Stress, Psychological , Animals , Dysbiosis/metabolism , Depression/metabolism , Depression/microbiology , Depression/psychology , Depression/etiology , Male , Humans , Stress, Psychological/metabolism , Stress, Psychological/microbiology , Stress, Psychological/psychology , Female , Adult , Mice , Restraint, Physical/psychology , Mice, Inbred C57BL , Gastrointestinal Microbiome , Brain-Gut Axis , Mouth/microbiology , Middle Aged , Saliva/metabolism , Saliva/microbiology , Behavior, Animal , Blood-Brain Barrier/metabolism
18.
Sci Rep ; 14(1): 10045, 2024 05 02.
Article En | MEDLINE | ID: mdl-38698013

Chronic stress has been implicated in mental illnesses and depressive behaviors. Somatostatin 4 receptor (SSTR4) has been shown to mediate anxiolytic and depression-like effects. Here, we aimed to explore the potential of SSTR4 as a diagnostic marker for chronic stress in mice. The mice were divided into single stress, chronic restraint stress, and control groups, and Sstr4 mRNA expression in the pituitary, lungs, and thymus, its protein expression in the thymus, were analyzed. Compared to controls, Sstr4 mRNA expression decreased significantly in the pituitary gland of the chronic and single-stress groups (P = 0.0181 and 0.0022, respectively) and lungs of the single-stress group (P = 0.0124), whereas it significantly increased in the thymus of the chronic-stress group (P = 0.0313). Thymic SSTR4 expression did not decrease significantly in stress groups compared to that in the control group (P = 0.0963). These results suggest that SSTR4 expression fluctuates in response to stress. Furthermore, Sstr4 mRNA expression dynamics in each organ differed based on single or chronic restraint stress-loading periods. In conclusion, this study suggests that investigating SSTR4 expression in each organ could allow for its use as a stress marker to estimate the stress-loading period and aid in diagnosing chronic stress.


Biomarkers , Receptors, Somatostatin , Stress, Psychological , Thymus Gland , Animals , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/genetics , Mice , Stress, Psychological/metabolism , Male , Biomarkers/metabolism , Thymus Gland/metabolism , Pituitary Gland/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Lung/metabolism , Chronic Disease , Stress, Physiological , Restraint, Physical
19.
FASEB J ; 38(10): e23684, 2024 May 31.
Article En | MEDLINE | ID: mdl-38795334

Exposure to chronic psychosocial stress is a risk factor for metabolic disorders. Because dipeptidyl peptidase-4 (DPP4) and cysteinyl cathepsin K (CTSK) play important roles in human pathobiology, we investigated the role(s) of DPP4 in stress-related adipocyte differentiation, with a focus on the glucagon-like peptide-1 (GLP-1)/adiponectin-CTSK axis in vivo and in vitro. Plasma and inguinal adipose tissue from non-stress wild-type (DPP4+/+), DPP4-knockout (DPP4-/-) and CTSK-knockout (CTSK-/-) mice, and stressed DPP4+/+, DPP4-/-, CTSK-/-, and DPP4+/+ mice underwent stress exposure plus GLP-1 receptor agonist exenatide loading for 2 weeks and then were analyzed for stress-related biological and/or morphological alterations. On day 14 under chronic stress, stress decreased the weights of adipose tissue and resulted in harmful changes in the plasma levels of DPP4, GLP-1, CTSK, adiponectin, and tumor necrosis factor-α proteins and the adipose tissue levels of CTSK, preadipocyte factor-1, fatty acid binding protein-4, CCAAT/enhancer binding protein-α, GLP-1 receptor, peroxisome proliferator-activated receptor-γ, perilipin2, secreted frizzled-related protein-4, Wnt5α, Wnt11 and ß-catenin proteins and/or mRNAs as well as macrophage infiltration in adipose tissue; these changes were rectified by DPP4 deletion. GLP-1 receptor activation and CTSK deletion mimic the adipose benefits of DPP4 deficiency. In vitro, CTSK silencing and overexpression respectively prevented and facilitated stress serum and oxidative stress-induced adipocyte differentiation accompanied with changes in the levels of pref-1, C/EBP-α, and PPAR-γ in 3T3-L1 cells. Thus, these findings indicated that increased DPP4 plays an essential role in stress-related adipocyte differentiation, possibly through a negative regulation of GLP-1/adiponectin-CTSK axis activation in mice under chronic stress conditions.


Adipocytes , Adiponectin , Cathepsin K , Cell Differentiation , Dipeptidyl Peptidase 4 , Glucagon-Like Peptide 1 , Mice, Knockout , Animals , Mice , Adiponectin/metabolism , Glucagon-Like Peptide 1/metabolism , Adipocytes/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl Peptidase 4/genetics , Cathepsin K/metabolism , Cathepsin K/genetics , Male , Mice, Inbred C57BL , Stress, Psychological/metabolism , 3T3-L1 Cells , Exenatide/pharmacology , PPAR gamma/metabolism , Adipogenesis
20.
Neuropharmacology ; 254: 109992, 2024 Aug 15.
Article En | MEDLINE | ID: mdl-38723742

Chronic primary pain, characterized by overlapping symptoms of chronic pain, anxiety, and depression, is strongly associated with stress and is particularly prevalent among females. Recent research has convincingly linked epigenetic modifications in the medial prefrontal cortex (mPFC) to chronic pain and chronic stress. However, our understanding of the role of histone demethylation in the mPFC in chronic stress-induced pain remains limited. In this study, we investigated the function of lysine-specific histone demethylase 1A (KDM1A/LSD1) in the context of chronic overlapping pain comorbid with anxiety and depression in female mice. We employed a chronic variable stress model to induce pain hypersensitivity in the face and hindpaws, as well as anxiety-like and depression-like behaviors, in female mice. Our findings revealed that chronic stress led to a downregulation of KDM1A mRNA and protein expression in the mPFC. Notably, overexpressing KDM1A in the mPFC alleviated the pain hypersensitivity, anxiety-like behaviors, and depression-like behaviors in female mice, without affecting basal pain responses or inducing emotional distress. Conversely, conditional knockout of KDM1A in the mPFC exacerbated pain sensitivity and emotional distress specifically in females. In summary, this study highlights the crucial role of KDM1A in the mPFC in modulating chronic stress-induced overlapping pain, anxiety, and depression in females. Our findings suggest that KDM1A may serve as a potential therapeutic target for treating chronic stress-related overlap pain and associated negative emotional disorders.


Chronic Pain , Down-Regulation , Histone Demethylases , Mice, Inbred C57BL , Prefrontal Cortex , Stress, Psychological , Animals , Prefrontal Cortex/metabolism , Female , Histone Demethylases/metabolism , Histone Demethylases/genetics , Stress, Psychological/metabolism , Stress, Psychological/psychology , Mice , Chronic Pain/metabolism , Chronic Pain/psychology , Depression/metabolism , Depression/etiology , Anxiety/metabolism , Mice, Knockout
...