Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.408
Filtrer
1.
Sci Rep ; 14(1): 13543, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866996

RÉSUMÉ

The objective of this study was to investigate spleen pathology and immune cell subset alterations in mice exposed to acute and chronic restraint stress over various timeframes. A deeper understanding of stress-induced spleen injuries can provide new insights into the mechanisms underlying stress-induced disorders. C57BL/6N mice were restrained for different durations (1, 3, 7, 14 and 21 days) for 6-8 h daily. The control mice were observed at the same time points. Post restraint, behavioural experiments were conducted to assess spleen weight, gross morphology and microscopic histological changes. Immunohistochemical staining was used to detect changes in glucocorticoid receptor (GR) expression, immune cell subsets and cell proliferation in response to stress. Our analysis revealed significant behavioural abnormalities in the stressed mice. In particular, there was an increase in the nuclear expression of GR beginning on Day 3, and it peaked on Day 14. The spleens of stressed mice displayed a reduction in size, disordered internal tissue structure and reduced cell proliferation. NK cells and M2-type macrophages exhibited immune cell subset alterations under stress, whereas T or B cells remained unaltered. Restraint stress can lead to pathomorphological alterations in spleen morphology, cell proliferation and immune cell counts in mice. These findings suggest that stress-induced pathological changes can disrupt immune regulation during stress.


Sujet(s)
Souris de lignée C57BL , Récepteurs aux glucocorticoïdes , Contention physique , Rate , Stress psychologique , Animaux , Rate/anatomopathologie , Rate/métabolisme , Récepteurs aux glucocorticoïdes/métabolisme , Souris , Mâle , Stress psychologique/immunologie , Prolifération cellulaire , Facteurs temps , Cellules tueuses naturelles/immunologie , Stress physiologique , Macrophages/immunologie , Macrophages/métabolisme
2.
Int Immunopharmacol ; 136: 112330, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38823180

RÉSUMÉ

An inflammatory response is one of the pathogeneses of depression. The anti-inflammatory and neuroprotective effects of auraptene have previously been confirmed. We established an inflammatory depression model by lipopolysaccharide (LPS) injection combined with unpredictable chronic mild stress (uCMS), aiming to explore the effects of auraptene on depressive-like behaviors in adult mice. Mice were divided into a control group, vehicle group, fluoxetine group, celecoxib group, and auraptene group. Then, behavioral tests were conducted to evaluate the effectiveness of auraptene in ameliorating depressive-like behavior. Cyclooxygenase-2 (COX-2), C-reactive protein (CRP), tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were examined by ELISA. Interleukin-10 (IL-10), interleukin-4 (IL-4), and transforming growth factor-ß (TGF-ß) were examined by protein chip technology. The morphology of microglia was observed by the immunohistochemical method. The data showed that, compared with the control group, the vehicle group mice exhibited a depressive-like behavioral phenotype, accompanied by an imbalance in inflammatory cytokines and the activation of microglia in the hippocampus. The depressive behaviors of the auraptene group's mice were significantly alleviated, along with the decrease in pro-inflammatory factors and increase in anti-inflammatory factors, while the activation of microglia was inhibited in the hippocampus. Subsequently, we investigated the role of auraptene in vitro-cultured BV-2 cells treated with LPS. The analysis showed that auraptene downregulated the expression of IL-6, TNF-α, and NO, and diminished the ratio of CD86/CD206. The results showed that auraptene reduced the excessive phagocytosis and ROS production of LPS-induced BV2 cells. In conclusion, auraptene relieved depressive-like behaviors in mice probably via modulating hippocampal neuroinflammation mediated by microglia.


Sujet(s)
Coumarines , Cytokines , Dépression , Hippocampe , Lipopolysaccharides , Microglie , Stress psychologique , Animaux , Microglie/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Mâle , Dépression/traitement médicamenteux , Dépression/immunologie , Dépression/induit chimiquement , Souris , Stress psychologique/traitement médicamenteux , Stress psychologique/immunologie , Coumarines/pharmacologie , Coumarines/usage thérapeutique , Cytokines/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Modèles animaux de maladie humaine , Comportement animal/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/immunologie , Souris de lignée C57BL , Médiateurs de l'inflammation/métabolisme
3.
J Integr Neurosci ; 23(5): 101, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38812387

RÉSUMÉ

The feeling of emotional tension, restlessness, pressure, and inability to relax is referred to as psychological stress. Although it is unclear how psychological stress affects neurobiological processes, several factors are thought to be involved, including central and peripheral neuroinflammation, structural degeneration in the prefrontal cortex and hippocampus, alterations in fear neurocircuitry, and neuroplasticity. Aside from data relating cognitive impairment to chronic low-grade inflammatory stress, there is growing evidence linking mental stress, oxidative stress, and systemic inflammation to the development of psychological disorders. After chronic and acute illnesses, insomnia, depression, anxiety, posttraumatic stress disorder, and cognitive impairment were reported. Cognitive impairment is exacerbated by systemic and central inflammatory processes. There is uncertainty about the potential mechanisms causing these symptoms, although they are likely complex, with systemic inflammation playing a significant role. Therefore, this review aims to investigate the role of inflammation in stress-induced cognitive impairment. Depicting the inflammatory mechanisms of cognitive impairment is critical for understanding and treating illnesses, such as chronic stress exposure and anxiety disorders.


Sujet(s)
Dysfonctionnement cognitif , Inflammation , Stress psychologique , Humains , Stress psychologique/physiopathologie , Stress psychologique/immunologie , Stress psychologique/complications , Dysfonctionnement cognitif/physiopathologie , Dysfonctionnement cognitif/étiologie , Inflammation/physiopathologie , Maladies neuro-inflammatoires/physiopathologie , Maladies neuro-inflammatoires/immunologie , Animaux
4.
Int J Mol Sci ; 25(10)2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38791301

RÉSUMÉ

Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries.


Sujet(s)
Inflammation , Granulocytes neutrophiles , Contention physique , Stress psychologique , Animaux , Souris , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Stress psychologique/complications , Stress psychologique/immunologie , Inflammation/anatomopathologie , Mâle , Souris de lignée C57BL , Pièges extracellulaires/métabolisme , Maladies gastro-intestinales/étiologie , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme
5.
Front Immunol ; 15: 1365871, 2024.
Article de Anglais | MEDLINE | ID: mdl-38756771

RÉSUMÉ

More than 20% of American adults live with a mental disorder, many of whom are treatment resistant or continue to experience symptoms. Other approaches are needed to improve mental health care, including prevention. The role of the microbiome has emerged as a central tenet in mental and physical health and their interconnectedness (well-being). Under normal conditions, a healthy microbiome promotes homeostasis within the host by maintaining intestinal and brain barrier integrity, thereby facilitating host well-being. Owing to the multidirectional crosstalk between the microbiome and neuro-endocrine-immune systems, dysbiosis within the microbiome is a main driver of immune-mediated systemic and neural inflammation that can promote disease progression and is detrimental to well-being broadly and mental health in particular. In predisposed individuals, immune dysregulation can shift to autoimmunity, especially in the presence of physical or psychological triggers. The chronic stress response involves the immune system, which is intimately involved with the gut microbiome, particularly in the process of immune education. This interconnection forms the microbiota-gut-immune-brain axis and promotes mental health or disorders. In this brief review, we aim to highlight the relationships between stress, mental health, and the gut microbiome, along with the ways in which dysbiosis and a dysregulated immune system can shift to an autoimmune response with concomitant neuropsychological consequences in the context of the microbiota-gut-immune-brain axis. Finally, we aim to review evidenced-based prevention strategies and potential therapeutic targets.


Sujet(s)
Axe cerveau-intestin , Encéphale , Dysbiose , Microbiome gastro-intestinal , Troubles mentaux , Santé mentale , Stress psychologique , Humains , Microbiome gastro-intestinal/immunologie , Axe cerveau-intestin/immunologie , Stress psychologique/immunologie , Stress psychologique/microbiologie , Dysbiose/immunologie , Troubles mentaux/immunologie , Troubles mentaux/microbiologie , Encéphale/immunologie , Animaux , Neuro-immunomodulation
6.
Breast Dis ; 43(1): 119-126, 2024.
Article de Anglais | MEDLINE | ID: mdl-38758989

RÉSUMÉ

INTRODUCTION: Housewives are a population at high risk of breast cancer due to repeated or chronic exposure to stress. Prevention in a simple yet evidence-based manner is needed. METHODS: This study is a narrative review of the potential of massage as breast cancer prevention through stress and immune system mechanisms. RESULTS: Massage is able to prevent chronic stress through improved sleep and fatigue and lower stress levels. Prevention of chronic stress will maximize the function of cells that eliminate cancer cells, such as B cells, T cells, and natural killer (NK) cells, and improve the balance of Foxp3 Tregulator cells. Partnered delivery massage will bring effective benefits for stress reduction. CONCLUSIONS: Massage can provide indirect prevention of breast cancer, and partnered delivery massage can be a good choice to reduce stress.


Sujet(s)
Tumeurs du sein , Massage , Stress psychologique , Humains , Tumeurs du sein/prévention et contrôle , Tumeurs du sein/immunologie , Massage/méthodes , Femelle , Stress psychologique/prévention et contrôle , Stress psychologique/immunologie , Système immunitaire , Cellules tueuses naturelles/immunologie
7.
Brain Behav Immun ; 119: 734-740, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38701886

RÉSUMÉ

The impact of psychological stress on physiological systems has been a focus of extensive research, particularly in understanding its diverse effects on immune system activity and disease risk. This meta-analysis explores the dynamic effect of acute stress on salivary immunoglobulin-A (S-IgA) levels, a key biomarker for secretory immunity within the oral environment. Analyzing data from 34 samples comprising 87 effect sizes and a total of 1,025 subjects, a multi-level approach is employed to account for the temporal variability in measuring the stress response. The results reveal a significant increase in S-IgA levels peaking around 10 min after stress exposure, followed by a return to baseline levels approximately 30 min later. In addition, the meta-analysis identified several research gaps of the extant literature, such as limitations in the considered time lag after stress. In conclusion, the findings emphasize the temporal nuances of the S-IgA response to stress, which can help to infer potential biological pathways and guide sampling designs in future studies. Further, we highlight the use of a multi-level meta-analysis approach to investigate the temporal dependencies of the interplay between stress and immune functioning.


Sujet(s)
Salive , Stress psychologique , Humains , Salive/immunologie , Salive/composition chimique , Salive/métabolisme , Stress psychologique/immunologie , Stress psychologique/métabolisme , Immunoglobuline A/métabolisme , Facteurs temps , Immunoglobuline A sécrétoire/métabolisme , Marqueurs biologiques/métabolisme , Femelle , Mâle , Adulte
8.
Brain Behav Immun ; 119: 898-907, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38718908

RÉSUMÉ

Marital quality shares ties to inflammatory conditions like cardiovascular disease and diabetes. For decades, research has focused on marital conflict as a primary mechanism given its potential to trigger inflammatory responses. However, longitudinal evidence suggests that marital conflict declines over time, and little attention has been paid to the inflammatory aftermath of other types of marital exchanges. A spouse's emotional distress is an important but overlooked marital context, as partners are exposed to each other's upsetting emotions throughout adulthood. To directly compare reactivity in proinflammatory gene expression to these two marital stressors and to examine differences by age and marital satisfaction, 203 community adults ages 25-90 (N = 102 couples) provided blood samples and rated their negative mood before and after they 1) watched their partner relive an upsetting personal memory and, in a separate visit 1-2 weeks later, 2) discussed a conflictual topic in their relationship. Controlling for age, sex, race/ethnicity, BMI, alcohol use, smoking, and comorbidities, increases in proinflammatory gene expression were significantly larger after the partner's upsetting disclosure than after marital conflict (B = 0.073, SE = 0.031, p = .018). This pattern paralleled emotional reactivity to the tasks, wherein negative mood rose more in response to the partner's disclosure than to marital conflict (B = 4.305, SE = 1.468, p = .004). In sum, proinflammatory and mood reactivity to spousal distress exceeded reactivity to marital conflict, a well-established marital stressor. Findings reveal spousal distress as a novel mechanism that may link marriage to inflammation-related diseases, and even pose risks for both happy and unhappy couples across adulthood.


Sujet(s)
Conflit familial , Inflammation , Mariage , Conjoints , Stress psychologique , Humains , Femelle , Mâle , Adulte d'âge moyen , Conflit familial/psychologie , Adulte , Sujet âgé , Conjoints/psychologie , Stress psychologique/psychologie , Stress psychologique/immunologie , Mariage/psychologie , Inflammation/immunologie , Inflammation/psychologie , Sujet âgé de 80 ans ou plus , Satisfaction personnelle , Émotions/physiologie , Détresse psychologique , Affect/physiologie
9.
J Affect Disord ; 359: 333-341, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38801920

RÉSUMÉ

BACKGROUND: Neuroimmune plays an important role in major depressive disorders (MDD). N-linked protein glycosylation (NLG) might contribute to depression by regulating the neuroinflammatory response. As microglia is the main executor of neuroimmune function in the central neural system (CNS), targeting the process of N-linked protein glycosylation of microglia in the mice used for studying depression might potentially offer new avenues for the strategy for MDD. METHODS: The chronic unpredictable mild stress (CUMS) mouse model was established for the whole brain microglia isolating. Then, RNA samples of microglia were extracted for transcriptome sequencing and mRNA analysis. Immunofluorescence (IF) was used to identify the expression level of NLG-related enzyme, B4galt1, in microglia. RESULTS: The data showed that NLG was positively related to depression. Moreover, the NLG-related gene, B4galt1 increased expression in the microglia of CUMS mice. Then, the inhibition of NLG reversed the depressive behavior in CUMS mice. The expression level of B4galt1 in CUMS mice was upregulating following the NLG-inhibitor treatment. Similar results haven't been observed in neurons. Information obtained from these experiments showed increasing expression of B4galt1 in microglia following depressive-like behaviors. CONCLUSIONS: These findings indicate that NLG in microglia is associated with MDD, and suggest that therapeutically targeting NLG might be an effective strategy for depression. LIMITATIONS: How to modulate the B4galt1 or NLG pathways in microglia efficiently and economically request new technologies.


Sujet(s)
Trouble dépressif majeur , Modèles animaux de maladie humaine , Microglie , Animaux , Souris , Microglie/métabolisme , Glycosylation , Trouble dépressif majeur/métabolisme , Mâle , Stress psychologique/métabolisme , Stress psychologique/immunologie , Dépression/métabolisme , Galactosyltransferases/génétique , Galactosyltransferases/métabolisme , Souris de lignée C57BL , Encéphale/métabolisme
10.
Int J Mol Sci ; 25(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38791423

RÉSUMÉ

The relationship between psychological stress, altered skin immunity, and autophagy-related genes (ATGs) is currently unclear. Psoriasis is a chronic skin inflammation of unclear etiology that is characterized by persistence and recurrence. Immune dysregulation and emotional disturbances are recognized as significant risk factors. Emerging clinical evidence suggests a possible connection between anxiety disorders, heightened immune system activation, and altered skin immunity, offering a fresh perspective on the initiation of psoriasis. The aim of this study was to explore the potential shared biological mechanisms underlying the comorbidity of psoriasis and anxiety disorders. Psoriasis and anxiety disorders data were obtained from the GEO database. A list of 3254 ATGs was obtained from the public database. Differentially expressed genes (DEGs) were obtained by taking the intersection of DEGs between psoriasis and anxiety disorder samples and the list of ATGs. Five machine learning algorithms used screening hub genes. The ROC curve was performed to evaluate diagnostic performance. Then, GSEA, immune infiltration analysis, and network analysis were carried out. The Seurat and Monocle algorithms were used to depict T-cell evolution. Cellchat was used to infer the signaling pathway between keratinocytes and immune cells. Four key hub genes were identified as diagnostic genes related to psoriasis autophagy. Enrichment analysis showed that these genes are indeed related to T cells, autophagy, and immune regulation, and have good diagnostic efficacy validated. Using single-cell RNA sequencing analysis, we expanded our understanding of key cellular participants, including inflammatory keratinocytes and their interactions with immune cells. We found that the CASP7 gene is involved in the T-cell development process, and correlated with γδ T cells, warranting further investigation. We found that anxiety disorders are related to increased autophagy regulation, immune dysregulation, and inflammatory response, and are reflected in the onset and exacerbation of skin inflammation. The hub gene is involved in the process of immune signaling and immune regulation. The CASP7 gene, which is related with the development and differentiation of T cells, deserves further study. Potential biomarkers between psoriasis and anxiety disorders were identified, which are expected to aid in the prediction of disease diagnosis and the development of personalized treatments.


Sujet(s)
Troubles anxieux , Autophagie , Biologie informatique , Apprentissage machine , Psoriasis , Analyse sur cellule unique , Stress psychologique , Psoriasis/génétique , Psoriasis/immunologie , Humains , Autophagie/génétique , Biologie informatique/méthodes , Stress psychologique/génétique , Stress psychologique/immunologie , Troubles anxieux/génétique , Réseaux de régulation génique , Analyse de profil d'expression de gènes , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie
12.
Behav Pharmacol ; 35(4): 211-226, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38651984

RÉSUMÉ

Stimulation of the innate immune system prior to stress exposure is a possible strategy to prevent depression under stressful conditions. Based on the innate immune system stimulating activities of zymosan A, we hypothesize that zymosan A may prevent the development of chronic stress-induced depression-like behavior. Our results showed that a single injection of zymosan A 1 day before stress exposure at a dose of 2 or 4 mg/kg, but not at a dose of 1 mg/kg, prevented the development of depression-like behaviors in mice treated with chronic social defeat stress (CSDS). The prophylactic effect of a single zymosan A injection (2 mg/kg) on CSDS-induced depression-like behaviors disappeared when the time interval between zymosan A and stress exposure was extended from 1 day or 5 days to 10 days, which was rescued by a second zymosan A injection 10 days after the first zymosan A injection and 4 days (4×, once daily) of zymosan A injections 10 days before stress exposure. Further analysis showed that a single zymosan A injection (2 mg/kg) 1 day before stress exposure could prevent the CSDS-induced increase in pro-inflammatory cytokines in the hippocampus and prefrontal cortex. Inhibition of the innate immune system by pretreatment with minocycline (40 mg/kg) abolished the preventive effect of zymosan A on CSDS-induced depression-like behaviors and CSDS-induced increase in pro-inflammatory cytokines in the brain. These results suggest that activation of the innate immune system triggered by zymosan A prevents the depression-like behaviors and neuroinflammatory responses in the brain induced by chronic stress.


Sujet(s)
Dépression , Hippocampe , Stress psychologique , Zymosan , Animaux , Zymosan/pharmacologie , Souris , Stress psychologique/immunologie , Mâle , Dépression/traitement médicamenteux , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Cytokines/métabolisme , Comportement animal/effets des médicaments et des substances chimiques , Défaite sociale , Immunisation/méthodes , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/immunologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Minocycline/pharmacologie , Relation dose-effet des médicaments
13.
Int Immunopharmacol ; 132: 111942, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38565045

RÉSUMÉ

Endometriosis (EM) is a gynecological inflammatory disease often accompanied by stress, chronic pelvic pain (CPP), anxiety, and depression, leading to a diminished quality of life. This review aims to discuss the relationship between systemic and local inflammatory responses in the central nervous system (CNS), focusing on glial dysfunctions (astrocytes and microglia) as in critical brain regions involved in emotion, cognition, pain processing, anxiety, and depression. The review presents that EM is connected to increased levels of pro-inflammatory cytokines in the circulation. Additionally, chronic stress and CPP as stressors may contribute to the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, depleting the production of inflammatory mediators in the circulatory system and the brain. The systemic cytokines cause blood-brain barrier (BBB) breakdown, activate microglia in the brain, and lead to neuroinflammation. Furthermore, CPP may induce neuronal morphological alterations in critical regions through central sensitization and the activation of glial cells. The activation of glial cells, particularly the polarization of microglia, leads to the activation of the NLRP3 inflammasome and the overproduction of inflammatory cytokines. These inflammatory cytokines interact with the signaling pathways involved in neural plasticity. Additionally, persistent inflammatory conditions in the brain lead to neuronal death, which is correlated with a reduced volume of key brain regions such as the hippocampus. This review highlights the involvement of glial cells in the pathogenesis of the mental comorbidities of EM (i.e., pain, anxiety, and depression) and to discuss potential therapeutic approaches for targeting the inflammation and activation of microglia in key brain regions.


Sujet(s)
Anxiété , Dépression , Endométriose , Névroglie , Humains , Femelle , Endométriose/immunologie , Endométriose/anatomopathologie , Dépression/immunologie , Dépression/étiologie , Dépression/métabolisme , Anxiété/immunologie , Animaux , Névroglie/immunologie , Inflammation/immunologie , Stress psychologique/immunologie , Cytokines/métabolisme , Encéphale/immunologie , Encéphale/anatomopathologie , Encéphale/métabolisme
15.
Brain Behav Immun ; 119: 572-596, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38663771

RÉSUMÉ

Chronic stress enhances the risk of neuropsychiatric disorders and contributes to the aggravation and chronicity of pain. The development of stress-associated diseases, including pain, is affected by individual vulnerability or resilience to stress, although the mechanisms remain elusive. We used the repeated social defeat stress model promoting susceptible and resilient phenotypes in male and female mice and induced knee mono-arthritis to investigate the impact of stress vulnerability on pain and immune system regulation. We analyzed different pain-related behaviors, measured blood cytokine and immune cell levels, and performed histological analyses at the knee joints and pain/stress-related brain areas. Stress susceptible male and female mice showed prolonged arthritis-associated hypersensitivity. Interestingly, hypersensitivity was exacerbated in male but not female mice. In males, stress promoted transiently increased neutrophils and Ly6Chigh monocytes, lasting longer in susceptible than resilient mice. While resilient male mice displayed persistently increased levels of the anti-inflammatory interleukin (IL)-10, susceptible mice showed increased levels of the pro-inflammatory IL-6 at the early- and IL-12 at the late arthritis stage. Although joint inflammation levels were comparable among groups, macrophage and neutrophil infiltration was higher in the synovium of susceptible mice. Notably, only susceptible male mice, but not females, presented microgliosis and monocyte infiltration in the prefrontal cortex at the late arthritis stage. Blood Ly6Chigh monocyte depletion during the early inflammatory phase abrogated late-stage hypersensitivity and the associated histological alterations in susceptible male mice. Thus, recruitment of blood Ly6Chigh monocytes during the early arthritis phase might be a key factor mediating the persistence of arthritis pain in susceptible male mice. Alternative neuro-immune pathways that remain to be explored might be involved in females.


Sujet(s)
Défaite sociale , Stress psychologique , Animaux , Mâle , Femelle , Souris , Stress psychologique/complications , Stress psychologique/immunologie , Stress psychologique/métabolisme , Souris de lignée C57BL , Cytokines/métabolisme , Arthrite/immunologie , Arthrite/métabolisme , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Cortex préfrontal/métabolisme , Hypersensibilité/immunologie , Hypersensibilité/métabolisme , Inflammation/métabolisme , Inflammation/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Douleur/métabolisme , Monocytes/métabolisme , Monocytes/immunologie , Encéphale/métabolisme , Encéphale/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Modèles animaux de maladie humaine , Facteurs sexuels
16.
Brain Behav Immun ; 119: 724-733, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38663776

RÉSUMÉ

Elucidating mechanisms by which early-life adversity (ELA) contributes to increased disease risk is important for mitigating adverse health outcomes. Prior work has found differences in immune cell gene expression related to inflammation and mitochondrial activity. Using a within-person between-group experimental design, we investigated differences in gene expression clusters across acute psychosocial stress and no-stress conditions. Participants were young adults (N = 29, aged 18 - 25 years, 62 % female, 47 % with a history of ELA). Gene expression was assessed in peripheral blood mononuclear cells collected at 8 blood draws spanning two 5-hour sessions (stress vs. no-stress) separated by a week, 4 across each session (number of observations = 221). We applied two unsupervised gene clustering methods - latent profile analysis (LPA) and weighted gene co-expression analysis (WGCNA) - to cluster genes with similar expression patterns across participants. LPA identified 11 clusters, 7 of which were significantly associated with ELA-status. WGCNA identified 5 clusters, 3 of which were significantly associated with ELA-status. LPA- and WGCNA-identified clusters were correlated, and all clusters were highly preserved across sessions and time. There was no significant effect of acute stress on cluster gene expression, but there was a significant effect of time, and significant differences by ELA-status. ELA-associated clusters related to RNA splicing/processing, inflammation, leukocyte differentiation and division, and mitochondrial activity were differentially expressed across time: ELA-exposed individuals showed decreased expression of these clusters at 90-minutes while controls showed increased expression. Our findings replicate previous work in this area and highlight additional mechanisms by which ELA may contribute to disease risk.


Sujet(s)
Expériences défavorables de l'enfance , Agranulocytes , Stress psychologique , Humains , Femelle , Stress psychologique/métabolisme , Stress psychologique/génétique , Stress psychologique/immunologie , Mâle , Adulte , Jeune adulte , Adolescent , Agranulocytes/métabolisme , Analyse de regroupements , Expression des gènes/génétique , Transcriptome , Inflammation/génétique , Inflammation/métabolisme
17.
Brain Behav Immun ; 119: 507-519, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38643954

RÉSUMÉ

Multiple myeloma (MM) is an incurable cancer and is the leading indication for autologous hematopoietic stem cell transplantation (HSCT). To be eligible for HSCT, a patient must have a caregiver, as caregivers play a central role in HSCT preparation and recovery. MM patients remain on treatment indefinitely, and thus patients and their caregivers face long-term challenges including the intensity of HSCT and perpetual therapy after transplant. Importantly, both patients and their caregivers show heightened depressive and anxiety symptoms, with dyadic correspondence evidenced and caregivers' distress often exceeding that of patients. An extensive psychoneuroimmunology (PNI) literature links distress with health via immune and neuroendocrine dysregulation as well as biological aging. However, data on PNI in the context of multiple myeloma - in patients or caregivers - are remarkably limited. Distress in MM patients has been associated with poorer outcomes including higher inflammation, greater one year post-HSCT hospital readmissions, and worse overall survival. Further, anxiety and depression are linked to biological aging and may contribute to the poor long-term health of both patients and caregivers. Because MM generally affects older adults, individual differences in biological aging may represent an important modifier of MM biology and HSCT treatment outcomes. There are a number of clinical scenarios in which biologically younger people could be prescribed more intensive therapies, with potential for greater benefit, by using a personalized cancer therapy approach based on the quantification of physiologic reserve. Further, despite considerable psychological demands, the effects of distress on health among MM caregivers is largely unexamined. Within this context, the current critical review highlights gaps in knowledge at the intersection of HSCT, inflammation, and biological aging in the context of MM. Research in this area hold promise for opportunities for novel and impactful psychoneuroimmunology (PNI) research to enhance health outcomes, quality of life, and longevity among both MM patients and their caregivers.


Sujet(s)
Anxiété , Aidants , Dépression , Transplantation de cellules souches hématopoïétiques , Myélome multiple , Psycho-neuro-immunologie , Transplantation autologue , Humains , Transplantation de cellules souches hématopoïétiques/psychologie , Transplantation de cellules souches hématopoïétiques/méthodes , Myélome multiple/immunologie , Myélome multiple/psychologie , Myélome multiple/thérapie , Aidants/psychologie , Dépression/immunologie , Dépression/psychologie , Stress psychologique/immunologie , Stress psychologique/psychologie , Vieillissement/immunologie , Vieillissement/psychologie , Qualité de vie/psychologie
19.
Cancer Immunol Res ; 12(5): 516-529, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38437646

RÉSUMÉ

As understanding of cancer has deepened, increasing attention has been turned to the roles of psychological factors, especially chronic stress-induced depression, in the occurrence and development of tumors. However, whether and how depression affects the progression of gliomas are still unclear. In this study, we have revealed that chronic stress inhibited the recruitment of tumor-associated macrophages (TAM) and other immune cells, especially M1-type TAMs and CD8+ T cells, and decreased the level of proinflammatory cytokines in gliomas, leading to an immunosuppressive microenvironment and glioma progression. Mechanistically, by promoting the secretion of stress hormones, chronic stress inhibited the secretion of the chemokine CCL3 and the recruitment of M1-type TAMs in gliomas. Intratumoral administration of CCL3 reprogrammed the immune microenvironment of gliomas and abolished the progression of gliomas induced by chronic stress. Moreover, levels of CCL3 and M1-type TAMs were decreased in the tumor tissues of glioma patients with depression, and CCL3 administration enhanced the antitumor effect of anti-PD-1 therapy in orthotopic models of gliomas undergoing chronic stress. In conclusion, our study has revealed that chronic stress exacerbates the immunosuppressive microenvironment and progression of gliomas by reducing the secretion of CCL3. CCL3 alone or in combination with an anti-PD-1 may be an effective immunotherapy for the treatment of gliomas with depression. See related Spotlight by Cui and Kang, p. 514.


Sujet(s)
Chimiokine CCL3 , Évolution de la maladie , Gliome , Stress psychologique , Microenvironnement tumoral , Animaux , Humains , Mâle , Souris , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Chimiokine CCL3/métabolisme , Gliome/immunologie , Gliome/métabolisme , Gliome/anatomopathologie , Gliome/traitement médicamenteux , Souris de lignée C57BL , Stress psychologique/immunologie , Stress psychologique/complications , Microenvironnement tumoral/immunologie , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme
20.
Nature ; 626(8001): 1108-1115, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38326622

RÉSUMÉ

Psychosocial stress has profound effects on the body, including the immune system and the brain1,2. Although a large number of pre-clinical and clinical studies have linked peripheral immune system alterations to stress-related disorders such as major depressive disorder (MDD)3, the underlying mechanisms are not well understood. Here we show that expression of a circulating myeloid cell-specific proteinase, matrix metalloproteinase 8 (MMP8), is increased in the serum of humans with MDD as well as in stress-susceptible mice following chronic social defeat stress (CSDS). In mice, we show that this increase leads to alterations in extracellular space and neurophysiological changes in the nucleus accumbens (NAc), as well as altered social behaviour. Using a combination of mass cytometry and single-cell RNA sequencing, we performed high-dimensional phenotyping of immune cells in circulation and in the brain and demonstrate that peripheral monocytes are strongly affected by stress. In stress-susceptible mice, both circulating monocytes and monocytes that traffic to the brain showed increased Mmp8 expression following chronic social defeat stress. We further demonstrate that circulating MMP8 directly infiltrates the NAc parenchyma and controls the ultrastructure of the extracellular space. Depleting MMP8 prevented stress-induced social avoidance behaviour and alterations in NAc neurophysiology and extracellular space. Collectively, these data establish a mechanism by which peripheral immune factors can affect central nervous system function and behaviour in the context of stress. Targeting specific peripheral immune cell-derived matrix metalloproteinases could constitute novel therapeutic targets for stress-related neuropsychiatric disorders.


Sujet(s)
Trouble dépressif majeur , Matrix metalloproteinase 8 , Monocytes , Stress psychologique , Animaux , Humains , Souris , Trouble dépressif majeur/sang , Trouble dépressif majeur/enzymologie , Trouble dépressif majeur/génétique , Trouble dépressif majeur/métabolisme , Espace extracellulaire/métabolisme , Matrix metalloproteinase 8/sang , Matrix metalloproteinase 8/déficit , Matrix metalloproteinase 8/génétique , Matrix metalloproteinase 8/métabolisme , Souris de lignée C57BL , Monocytes/composition chimique , Monocytes/immunologie , Monocytes/métabolisme , Noyau accumbens/métabolisme , Noyau accumbens/anatomopathologie , Tissu parenchymateux/métabolisme , Analyse de l'expression du gène de la cellule unique , Comportement social , Isolement social , Stress psychologique/sang , Stress psychologique/génétique , Stress psychologique/immunologie , Stress psychologique/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...