Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 957
Filter
Add more filters

Publication year range
1.
Immunity ; 54(2): 225-234.e6, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33476547

ABSTRACT

Microglia are activated in many neurological diseases and have been suggested to play an important role in the development of affective disorders including major depression. To investigate how microglial signaling regulates mood, we used bidirectional chemogenetic manipulations of microglial activity in mice. Activation of microglia in the dorsal striatum induced local cytokine expression and a negative affective state characterized by anhedonia and aversion, whereas inactivation of microglia blocked aversion induced by systemic inflammation. Interleukin-6 signaling and cyclooxygenase-1 mediated prostaglandin synthesis in the microglia were critical for the inflammation-induced aversion. Correspondingly, microglial activation led to a prostaglandin-dependent reduction of the excitability of striatal neurons. These findings demonstrate a mechanism by which microglial activation causes negative affect through prostaglandin-dependent modulation of striatal neurons and indicate that interference with this mechanism could milden the depressive symptoms in somatic and psychiatric diseases involving microglial activation.


Subject(s)
Anhedonia/physiology , Corpus Striatum/immunology , Depression/immunology , Microglia/immunology , Neurons/physiology , Animals , Animals, Genetically Modified , Behavior, Animal , Cells, Cultured , Disease Models, Animal , Humans , Inflammation , Interleukin-6/metabolism , Macrophage Activation , Mice , Neurogenic Inflammation , Prostaglandins/metabolism
2.
Pharmacol Rev ; 76(5): 956-969, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39084934

ABSTRACT

Depression is a highly prevalent disorder and a leading cause of disability worldwide. It has a major impact on the affected individual and on society as a whole. Regrettably, current available treatments for this condition are insufficient in many patients. In recent years, the gut microbiome has emerged as a promising alternative target for treating and preventing depressive disorders. However, the microbes that form this ecosystem do not act alone but are part of a complicated network connecting the gut and the brain that influences our mood. Host cells that are in intimate contact with gut microbes, such as the epithelial cells forming the gut barrier and the immune cells in their vicinity, play a key role in the process. These cells continuously shape immune responses to maintain healthy communication between gut microbes and the host. In this article, we review how the interplay among epithelial cells, the immune system, and gut microbes mediates gut-brain communication to influence mood. We also discuss how advances in our knowledge of the mechanisms underlying the gut-brain axis could contribute to addressing depression. SIGNIFICANCE STATEMENT: This review does not aim to systematically describe intestinal microbes that might be beneficial or detrimental for depression. We have adopted a novel point of view by focusing on potential mechanisms underlying the crosstalk between gut microbes and their intestinal environment to control mood. These pathways could be targeted by well defined and individually tailored dietary interventions, microbes, or microbial metabolites to ameliorate depression and decrease its important social and economic impact.


Subject(s)
Brain-Gut Axis , Depression , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Animals , Brain-Gut Axis/physiology , Depression/immunology , Depression/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism
3.
Am J Physiol Cell Physiol ; 327(1): C205-C212, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38826138

ABSTRACT

Major depressive disorder (MDD) affects millions of individuals worldwide, leading to considerable social and economic costs. Despite advancements in pharmacological treatments, achieving remission remains a key challenge, with a substantial number of patients showing resistance to existing therapies. This resistance is often associated with elevated levels of proinflammatory cytokines, suggesting a connection between inflammation, MDD pathophysiology, and treatment efficacy. The observation of increased immune activation in about a quarter of patients with MDD resulted in the distinction between inflammatory and noninflammatory endotypes. Although anti-inflammatory treatments show promise in alleviating depression-like symptoms, responses are heterogeneous, thus highlighting the importance of identifying distinct inflammatory endotypes to tailor effective therapeutic strategies. The intestinal microbiome emerges as a crucial modulator of mental health, mediating its effects partially through different immune pathways. Microbiota-derived short-chain fatty acids (SCFAs) significantly impact innate and adaptive immune cells, regulating their differentiation, function, and cellular response. Furthermore, gut-educated immune cells reach the border regions of the central nervous system (CNS), regulating glial cell functions. Although the CNS modulates immune responses via efferent parts of the vagus nerve, afferent tracts concurrently transport information on peripheral inflammation back to the brain. This bidirectional communication is particularly relevant in depression, allowing for therapeutic stimulation of the vagus nerve in the context of inflammatory depression endotypes. In this review, we explore the intricate relationship between inflammation and depression, discuss how inflammatory signals are translated into depressive-like symptoms, and highlight immune-modulating therapeutic avenues.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Inflammation , Humans , Gastrointestinal Microbiome/immunology , Depressive Disorder, Major/immunology , Depressive Disorder, Major/diagnosis , Animals , Inflammation/immunology , Brain-Gut Axis/physiology , Cytokines/metabolism , Cytokines/immunology , Depression/immunology , Depression/diagnosis , Brain/immunology , Brain/physiopathology , Brain/metabolism
4.
BMC Immunol ; 25(1): 61, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333855

ABSTRACT

Major Depressive Disorder, or depression, has been extensively linked to dysregulated HPA axis function, chronic inflammation and cardiovascular diseases. While the former two have been studied in depth, the mechanistic connection between depression and cardiovascular disease is unclear. As major mediators of vascular homeostasis, vascular pathology and immune activity, endothelial cells represent an important player connecting the diseases. Exaggerated inflammation and glucocorticoid function are important topics to explore in the endothelial response to MDD. Glucocorticoid resistance in several cell types strongly promotes inflammatory signaling and results in worsened severity in many diseases. However, endothelial health and inflammation in chronic stress and depression are rarely considered from the perspective of glucocorticoid signaling and resistance. In this review, we aim to discuss (1) endothelial dysfunction in depression, (2) inflammation in depression, (3) general glucocorticoid resistance in depression and (4) endothelial glucocorticoid resistance in depression co-morbid inflammatory diseases. We will first describe vascular pathology, inflammation and glucocorticoid resistance separately in depression and then describe their potential interactions with one another in depression-relevant diseases. Lastly, we will hypothesize potential mechanisms by which glucocorticoid resistance in endothelial cells may contribute to vascular disease states in depressed people. Overall, endothelial-glucocorticoid signaling may play an important role in connecting depression and vascular pathology and warrants further study.


Subject(s)
Cardiovascular Diseases , Glucocorticoids , Inflammation , Humans , Cardiovascular Diseases/etiology , Inflammation/immunology , Animals , Signal Transduction , Endothelial Cells/metabolism , Depression/immunology , Depression/physiopathology , Endothelium, Vascular/physiopathology , Receptors, Glucocorticoid/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Depressive Disorder, Major/immunology
5.
J Neuroinflammation ; 21(1): 159, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898454

ABSTRACT

A significant decrease in estrogen levels puts menopausal women at high risk for major depression, which remains difficult to cure despite its relatively clear etiology. With the discovery of abnormally elevated inflammation in menopausal depressed women, immune imbalance has become a novel focus in the study of menopausal depression. In this paper, we examined the characteristics and possible mechanisms of immune imbalance caused by decreased estrogen levels during menopause and found that estrogen deficiency disrupted immune homeostasis, especially the levels of inflammatory cytokines through the ERα/ERß/GPER-associated NLRP3/NF-κB signaling pathways. We also analyzed the destruction of the blood-brain barrier, dysfunction of neurotransmitters, blockade of BDNF synthesis, and attenuation of neuroplasticity caused by inflammatory cytokine activity, and investigated estrogen-immuno-neuromodulation disorders in menopausal depression. Current research suggests that drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.


Subject(s)
Estrogens , Menopause , Humans , Female , Menopause/immunology , Menopause/metabolism , Estrogens/metabolism , Animals , Depression/immunology , Depression/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/physiology , Cytokines/metabolism
6.
J Med Virol ; 96(8): e29864, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39158051

ABSTRACT

Inflammation and autoimmune responses contribute to the pathophysiology of Long COVID, and its affective and chronic fatigue syndrome symptoms, labeled "the physio-affective phenome." To investigate whether Long COVID and its physio-affective phenome are linked to autoimmunity to the tight junction proteins, zonulin and occludin (ZOOC), and immune reactivity to lipopolysaccharides (LPS), and whether the latter are associated with signs of human herpes virus-6 (HHV-6) reactivation, autoimmunity directed against oligodendrocyte and neuronal proteins, including myelin basic protein. IgA/IgM/IgG responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), HHV-6, ZOOC, and neuronal proteins, C-reactive protein (CRP), and advanced oxidation protein products (AOPPs), were measured in 90 Long COVID patients and 90 healthy controls. The physio-affective phenome was conceptualized as a factor extracted from physical and affective symptom domains. Neural network identified IgA directed to LPS (IgA-LPS), IgG-ZOOC, IgG-LPS, and IgA-ZOOC as important variables associated with Long COVID diagnosis with an area under the ROC curve of 0.755. Partial Least Squares analysis showed that 40.9% of the variance in the physio-affective phenome was explained by CRP, IgA-myelin basic protein (MBP), and IgG-MBP. A large part of the variances in both autoimmune responses to MBP (36.3%-39.7%) was explained by autoimmunity (IgA and IgG) directed to ZOOC. The latter was strongly associated with indicants of HHV-6 reactivation, which in turn was associated with increased IgM-SARS-CoV-2. Autoimmunity against components of the tight junctions and increased bacterial translocation may be involved in the pathophysiology of Long COVID's physio-affective phenome.


Subject(s)
Autoimmunity , COVID-19 , Fatigue Syndrome, Chronic , Herpesvirus 6, Human , Inflammation , Tight Junctions , Humans , Fatigue Syndrome, Chronic/immunology , Fatigue Syndrome, Chronic/virology , Herpesvirus 6, Human/immunology , Female , Male , Middle Aged , Tight Junctions/immunology , COVID-19/immunology , Inflammation/immunology , Adult , Occludin , Depression/immunology , SARS-CoV-2/immunology , Aged , Immunoglobulin G/blood , Post-Acute COVID-19 Syndrome , Immunoglobulin A/blood , Lipopolysaccharides/immunology , Autoantibodies/blood , Autoantibodies/immunology , Antibodies, Viral/blood , Roseolovirus Infections/immunology , Roseolovirus Infections/complications , Roseolovirus Infections/virology , Haptoglobins , Protein Precursors
7.
Brain Behav Immun ; 120: 141-150, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38777289

ABSTRACT

BACKGROUND: Dysregulation of the immune system has been associated with psychiatric disorders and pregnancy-related complications, such as perinatal depression. However, the immune characteristics specific to perinatal anxiety remain poorly understood. In this study, our goal was to examine specific immune characteristics related to prenatal anxiety within the context of a randomized controlled trial designed to alleviate anxiety symptoms-the Happy Mother - Healthy Baby (HMHB) study in Rawalpindi, Pakistan. MATERIALS AND METHODS: Pregnant women (n = 117) were followed prospectively in the 1st, 2nd, and 3rd trimesters (T1, T2, T3) and at 6 weeks postpartum (PP6). Each visit included a blood draw and anxiety evaluation (as measured by the anxiety subscale of the Hospital Anxiety and Depression Scale - HADS -using a cutoff ≥ 8). We enrolled both healthy controls and participants with anxiety alone; those with concurrent depression were excluded. RESULTS: K-means cluster analysis revealed three anxiety clusters: Non-Anxiety, High and Consistent Anxiety, and Decreasing Anxiety. Principal components analysis revealed two distinct clusters of cytokine and chemokine activity. Women within the High and Consistent Anxiety group had significantly elevated chemokine activity across pregnancy (in trimester 1 (ß = 0.364, SE = 0.178, t = 2.040, p = 0.043), in trimester 2 (ß = 0.332, SE = 0.164, t = 2.020, p = 0.045), and trimester 3 (ß = 0.370, SE = 0.179, t = 2.070, p = 0.040) compared to Non-Anxiety group. Elevated chemokine activity was associated with low birthweight (LBW) and small for gestational age (SGA). CONCLUSION: Our findings reveal a unique pattern of immune dysregulation in pregnant women with anxiety in a Pakistani population and offer preliminary evidence that immune dysregulation associated with antenatal anxiety may be associated with birth outcomes. The dysregulation in this population is distinct from that in our other studies, indicating that population-level factors other than anxiety may play a substantial role in the differences found. (Clinicaltrials.gov # NCT04566861).


Subject(s)
Anxiety , Pregnancy Complications , Humans , Female , Pregnancy , Pakistan , Adult , Anxiety/immunology , Pregnancy Complications/immunology , Pregnancy Complications/psychology , Cytokines/blood , Behavior Therapy/methods , Young Adult , Chemokines/blood , Phenotype , Depression/immunology , Prospective Studies , Anxiety Disorders/immunology
8.
Brain Behav Immun ; 120: 403-412, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38871062

ABSTRACT

Persistent psychological stress can affect immune homeostasis and is a key factor in the development of depression. Many efforts are focused on the identifcation of pathways that link the immune system and mood disorders. Here, we found that psychological stress caused an increase in the frequency of brain-associated neutrophils and the level of neutrophil-specific antigen CD177 on peripheral neutrophils in male mice. Upregulated levels of blood CD177 are associated with depression in humans. Neutrophil depletion or Cd177 deficiency protected mice from stress-induced behavioral deficits. Importantly, adoptive transfer of CD177+ neutrophils from stressed mice increased the frequency of brain-associated leukocytes, including neutrophils, and caused behavioral defects in naive mice. These effects may be related to the endothelial adhesion advantage of CD177+ neutrophils and the interference of serine protease on endothelial junction. Our findings suggest a critical link between circulating CD177+ neutrophils and psychological stress-driven behavioral disorder.


Subject(s)
Behavior, Animal , Mice, Inbred C57BL , Neutrophils , Stress, Psychological , Animals , Humans , Male , Mice , Behavior, Animal/physiology , Brain/metabolism , Depression/metabolism , Depression/immunology , GPI-Linked Proteins/metabolism , Neutrophils/metabolism , Receptors, Cell Surface/metabolism , Stress, Psychological/metabolism , Stress, Psychological/immunology
9.
Brain Behav Immun ; 119: 507-519, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643954

ABSTRACT

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.


Subject(s)
Anxiety , Caregivers , Depression , Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Psychoneuroimmunology , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Transplantation/psychology , Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/immunology , Multiple Myeloma/psychology , Multiple Myeloma/therapy , Caregivers/psychology , Depression/immunology , Depression/psychology , Stress, Psychological/immunology , Stress, Psychological/psychology , Aging/immunology , Aging/psychology , Quality of Life/psychology
10.
Brain Behav Immun ; 121: 257-268, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089535

ABSTRACT

Major Depressive Disorder (MDD) is a heterogeneous disorder that affects twice as many women than men. Precluding advances in more tailored and efficacious treatments for depression is the lack of reliable biomarkers. While depression is linked to elevations in inflammatory immune system functioning, this relationship is not evident among all individuals with depression and may vary based on symptom subtypes and/or sex. This systematic review and meta-analysis examined whether inflammatory immune peripheral markers of depression are sex-specific. PRISMA guidelines were followed for the systematic review, and a comprehensive search strategy that identified studies from PubMed and PsycInfo was applied. Studies were included if they reported C-reactive protein (CRP), interleukin (IL)-6, tumor necrosis factor (TNF)-α and/or IL-1ß for males and/or females among depressed and healthy adults. We identified 23 studies that satisfied these inclusion criteria. Random-effects meta-analysis models were fit, and measures of association were summarized between levels of circulating markers of inflammation in depressed and healthy males and females. Sex-based analyses revealed elevated levels of CRP among females with depression (Cohen's d = 0.19) relative to their healthy counterparts (p = 0.02), an effect not apparent among males (Cohen's d = -0.01). Similarly, levels of IL-6 were increased among females with depression compared to healthy controls (Cohen's d = 0.51; p = 0.04), but once again this was not found among males (Cohen's d = 0.16). While TNF-α levels were elevated among individuals with depression compared to controls (p = 0.01), no statistically significant sex differences were found. The meta-analysis for IL-1ß resulted in only three articles, and thus, results are presented in the supplemental section. This meta-analysis advances our understanding of the unique involvement of inflammatory biomarkers in depression among men and women, which may help inform more tailored sex-specific treatment approaches in the future.


Subject(s)
Biomarkers , C-Reactive Protein , Depressive Disorder, Major , Inflammation , Sex Factors , Female , Humans , Male , Biomarkers/blood , Biomarkers/metabolism , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Depression/blood , Depression/immunology , Depression/metabolism , Depressive Disorder, Major/blood , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Inflammation/blood , Inflammation/metabolism , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
11.
Clin Exp Rheumatol ; 42(7): 1350-1358, 2024 07.
Article in English | MEDLINE | ID: mdl-38147314

ABSTRACT

OBJECTIVES: To explore the risk factors of anxiety and depression, especially their association with serum autoantibodies, in patients with connective tissue diseases (CTDs). METHODS: Three hundred and fifty-two inpatients with CTDs were recruited and their demographic, serological and imaging data were collected through the medical record system. Depression and anxiety were assessed by the Patient Health Questionnaire-9 (PHQ-9) and the Generalized Anxiety Disorder-7 Scale (GAD-7) respectively. Analysis of variance (ANOVA), rank sum test, chi-square test and logistic regression were performed to investigate risk factors for depression and anxiety. RESULTS: The prevalence of depression (PHQ-9 ≥ 5) and anxiety (GAD-7 ≥5) in CTD patients was significantly higher than that in the Chinese general population (depression: 44.3% vs. 32.2%, anxiety: 39.5% vs. 22.2%). Sleep time was a protective factor for both depression and anxiety (OR=0.734, 95% CI: 0.616~0.874, p<0.001 and OR=0.684, 95% CI: 0.559~0.835, P<0.001, respectively) while anti-Ro52 antibody was a risk factor for them (OR=5.466, 95% CI: 2.978~10.032, p<0.001 and OR=4.075, 95% CI: 2.073~8.010, p<0.001, respectively). Further analysis showed that anti-Ro52 antibody was a risk factor for depression and anxiety in all four subgroups, namely SLE, SS, RA, and other CTDs. CONCLUSIONS: Anti-Ro52 antibody is probably a risk factor for depression and anxiety in patients with connective tissue diseases. CTD patients with the presence of anti-Ro52 antibody are more prone to depression and anxiety than those without it.


Subject(s)
Anxiety , Connective Tissue Diseases , Depression , Ribonucleoproteins , Humans , Female , Male , Middle Aged , Connective Tissue Diseases/immunology , Connective Tissue Diseases/psychology , Connective Tissue Diseases/blood , Connective Tissue Diseases/epidemiology , Connective Tissue Diseases/diagnosis , Cross-Sectional Studies , Anxiety/epidemiology , Anxiety/immunology , Anxiety/psychology , Adult , Risk Factors , Depression/epidemiology , Depression/immunology , Depression/psychology , Ribonucleoproteins/immunology , Prevalence , China/epidemiology , Aged , Antibodies, Antinuclear/blood , Autoantibodies/blood , Biomarkers/blood , Logistic Models
12.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673781

ABSTRACT

Vagus nerve stimulation (VNS) represents a long-term adjunctive treatment option in patients with difficult-to-treat depression (DTD). Anti-inflammatory effects have been discussed as a key mechanism of action of VNS. However, long-term investigations in real-world patients are sparse. In this naturalistic observational study, we collected data on cytokines in peripheral blood in n = 6 patients (mean age 47.8) with DTD and VNS treatment at baseline and at 6 months follow-up. We have identified clusters of peripheral cytokines with a similar dynamic over the course of these 6 months using hierarchical clustering. We have investigated cytokine changes from baseline to 6 months as well as the relationship between the cytokine profile at 6 months and long-term response at 12 months. After 6 months of VNS, we observed significant correlations between cytokines (p < 0.05) within the identified three cytokine-pairs which were not present at baseline: IL(interleukin)-6 and IL-8; IL-1ß and TNF-α; IFN-α2 and IL-33. At 6 months, the levels of all the cytokines of interest had decreased (increased in non-responders) and were lower (5-534 fold) in responders to VNS than in non-responders: however, these results were not statistically significant. VNS-associated immunomodulation might play a role in long-term clinical response to VNS.


Subject(s)
Cytokines , Vagus Nerve Stimulation , Humans , Cytokines/blood , Cytokines/metabolism , Male , Female , Middle Aged , Vagus Nerve Stimulation/methods , Adult , Depression/therapy , Depression/immunology , Treatment Outcome , Immunomodulation
13.
Int J Mol Sci ; 25(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39337372

ABSTRACT

Maternal immune activation (MIA) during pregnancy has been increasingly recognized as a critical factor in the development of neurodevelopmental disorders, with potential sex-specific impacts that are not yet fully understood. In this study, we utilized a murine model to explore the behavioral and molecular consequences of MIA induced by lipopolysaccharide (LPS) administration on embryonic day 12.5. Our findings indicate that male offspring exposed to LPS exhibited significant increases in anxiety-like and depression-like behaviors, while female offspring did not show comparable changes. Molecular analyses revealed alterations in pro-inflammatory cytokine levels and synaptic gene expression in male offspring, suggesting that these molecular disruptions may underlie the observed behavioral differences. These results emphasize the importance of considering sex as a biological variable in studies of neurodevelopmental disorders and highlight the need for further molecular investigations to understand the mechanisms driving these sex-specific outcomes. Our study contributes to the growing evidence that prenatal immune challenges play a pivotal role in the etiology of neurodevelopmental disorders and underscores the potential for sex-specific preventative approaches of MIA.


Subject(s)
Behavior, Animal , Disease Models, Animal , Lipopolysaccharides , Neurodevelopmental Disorders , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Mice , Male , Neurodevelopmental Disorders/immunology , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/chemically induced , Prenatal Exposure Delayed Effects/immunology , Behavior, Animal/drug effects , Cytokines/metabolism , Anxiety/immunology , Sex Factors , Depression/immunology , Sex Characteristics , Mice, Inbred C57BL
14.
Brain Behav Immun ; 107: 414-418, 2023 01.
Article in English | MEDLINE | ID: mdl-36116693

ABSTRACT

BACKGROUND: Although factors associated with the antibody response to the BNT162b2 mRNA COVID-19 vaccine have been reported, psychological factors have not been examined. Depression or anxiety may affect vaccine reactions because these factors influence immune responses. This study aimed to determine whether psychological status at the time of vaccination predicts antibody responses. METHODS: A prospective observational study of the BNT162b2 mRNA COVID-19 vaccine response was carried out among individuals attending for an annual health check-up. Participants included 78 volunteers out of 80 hospital workers in Nagoya, Japan. No participants had been infected with COVID-19 and all gave written informed consent to participate in the study. Blood samples were obtained approximately 28 days after the second dose of the vaccine, and antibody titers of the SARS-CoV-2 spike protein were determined using the SARS-CoV-2 IgG II Quant assay. Participants completed the Japanese version of the hospital anxiety and depression scale (HADS) questionnaire, one day before both vaccinations. Participants also recorded any adverse reactions, such as body temperature and other side effects, every day for two weeks after each dose. The relationships between antibody titers and the predictive factors were analyzed using multiple linear regression analysis, with the antibody titers as the dependent variables, followed by univariate analysis. RESULTS: Multiple linear regression analysis revealed that no or excessive alcohol intake (p = 0.039), poor results from a health check-up (p = 0.011), a longer duration between the second dose and blood collection (p = 0.039), and increased degree of depressive symptoms (p = 0.041) were significant negative predictors of antibody titers, while body temperature one day after the second dose as a significant positive predictor of antibody titers (p < 0.0005). CONCLUSION: We identified that depressive symptoms just before the second dose of the BNT162b2 mRNA COVID-19 were an independent negative predictor of antibody responses, in addition to other factors. Our results highlight the importance of mental health at the time of vaccination to achieve the higher antibody responses necessary to acquire humoral immunity.


Subject(s)
COVID-19 Vaccines , COVID-19 , Depression , Humans , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Hospitals , SARS-CoV-2 , Depression/immunology , Antibodies, Viral/blood , Japan , Health Personnel
15.
J Neuroinflammation ; 19(1): 12, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34996472

ABSTRACT

BACKGROUND: Innate immune pre-stimulation can prevent the development of depression-like behaviors in chronically stressed mice; however, whether the same stimulation prevents the development of anxiety-like behaviors in animals remains unclear. We addressed this issue using monophosphoryl lipid A (MPL), a derivative of lipopolysaccharide (LPS) that lacks undesirable properties of LPS but still keeps immune-enhancing activities. METHODS: The experimental mice were pre-injected intraperitoneally with MPL before stress exposure. Depression was induced through chronic social defeat stress (CSDS). Behavioral tests were conducted to identify anxiety-like behaviors. Real-time polymerase chain reaction (PCR) and biochemical assays were employed to examine the gene and protein expression levels of pro-inflammatory markers. RESULTS: A single MPL injection at the dose of 400 and 800 µg/kg 1 day before stress exposure prevented CSDS-induced anxiety-like behaviors, and a single MPL injection (400 µg/kg) five but not 10 days before stress exposure produced similar effect. The preventive effect of MPL on anxiety-like behaviors was also observed in CSDS mice who received a second MPL injection 10 days after the first MPL injection or a 4 × MPL injection 10 days before stress exposure. MPL pre-injection also prevented the production of pro-inflammatory cytokines in the hippocampus and medial prefrontal cortex in CSDS mice, and inhibiting the central immune response by minocycline pretreatment abrogated the preventive effect of MPL on CSDS-induced anxiety-like behaviors and pro-inflammatory cytokine productions in the brain. CONCLUSIONS: Pre-stimulation of the innate immune system by MPL can prevent chronic stress-induced anxiety-like behaviors and neuroinflammatory responses in the brain in mice.


Subject(s)
Anxiety/immunology , Immunity, Innate/drug effects , Lipid A/analogs & derivatives , Prefrontal Cortex/drug effects , Social Defeat , Stress, Psychological/immunology , Animals , Depression/immunology , Lipid A/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Prefrontal Cortex/immunology , Social Behavior
16.
Brain Behav Immun ; 106: 21-29, 2022 11.
Article in English | MEDLINE | ID: mdl-35870669

ABSTRACT

BACKGROUND: Childhood trauma (CT) is robustly associated with psychiatric disorders including major depressive and anxiety disorders across the life span. The innate immune system may play a role in the relation between CT and stress-related psychopathology. However, whether CT influences the innate production capacity of cytokine levels following ex vivo stimulation by lipopolysaccharide (LPS), is currently unknown. METHODS: Using data from the Netherlands Study of Depression and Anxiety (NESDA, n=1237), we examined whether CT (emotional neglect, emotional, physical, and sexual abuse before the age of 16), assessed by the Childhood Trauma Interview, was associated with levels in supernatants of interferon (IFN)γ, interleukin-2 (IL-2), IL-4, IL-6, IL-8, IL-10, IL-18, monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein (MIP)-1α, MIP-1ß, matrix metalloproteinase-2 (MMP-2), TNFα and TNFß after ex vivo stimulation with LPS. Cytokines were analysed individually and cumulatively (overall inflammation index and number of cytokines in high-risk quartile (HRQ)) using linear regression analyses. RESULTS: After adjustment for demographic, lifestyle, and health-related covariates, total CT severity was associated with the overall inflammation index (ß = 0.085, PFDR = 0.011), the number of cytokines in HRQ (ß = 0.063, PFDR = 0.036), and individual markers of IL-2 (ß = 0.067, PFDR = 0.036), IL-6 (ß = 0.091 PFDR = 0.011), IL-8 (ß = 0.085 PFDR = 0.011), IL-10 (ß = 0.094 PFDR = 0.011), MCP-1 (ß = 0.081 PFDR = 0.011), MIP-1α (ß = 0.061 PFDR = 0.047), MIP1-ß (ß = 0.077 PFDR = 0.016), MMP-2 (ß = 0.070 PFDR = 0.027), and TNFß (ß = 0.078 PFDR = 0.016). Associations were strongest for individuals with severe CT, reporting multiple types or higher frequencies of trauma. Half of the findings persisted after adjustment for psychiatric status. The findings were consistent across different CT types. CONCLUSION: Childhood Trauma is associated with increased LPS-stimulated cytokine levels, with evidence for a dose-response relationship. Our results highlight a dysregulated innate immune system capacity in adults with CT, which could contribute to an increased vulnerability for psychopathology and somatic disorders across the lifespan.


Subject(s)
Adverse Childhood Experiences , Anxiety , Depression , Immunity, Innate , Adult , Anxiety/immunology , Anxiety Disorders/immunology , Chemokine CCL2 , Chemokine CCL3 , Chemokine CCL4 , Cytokines/metabolism , Depression/immunology , Depressive Disorder, Major/immunology , Humans , Inflammation , Interferons , Interleukin-10 , Interleukin-18 , Interleukin-2 , Interleukin-4 , Interleukin-6 , Interleukin-8 , Lipopolysaccharides , Matrix Metalloproteinase 2 , Netherlands/epidemiology , Tumor Necrosis Factor-alpha
17.
Brain ; 144(3): 746-760, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33279966

ABSTRACT

Major depression is a prevalent illness that increases the risk of several neurological conditions. These include stroke, cardiovascular disease, and dementia including Alzheimer's disease. In this review we ask whether certain types of depression and associated loneliness may be a harbinger of cognitive decline and possibly even dementia. We propose that chronic stress and inflammation combine to compromise vascular and brain function. The resulting increases in proinflammatory cytokines and microglial activation drive brain pathology leading to depression and mild cognitive impairment, which may progress to dementia. We present evidence that by treating the inflammatory changes, depression can be reversed in many cases. Importantly, there is evidence that anti-inflammatory and antidepressant treatments may reduce or prevent dementia in people with depression. Thus, we propose a model in which chronic stress and inflammation combine to increase brain permeability and cytokine production. This leads to microglial activation, white matter damage, neuronal and glial cell loss. This is first manifest as depression and mild cognitive impairment, but can eventually evolve into dementia. Further research may identify clinical subgroups with inflammatory depression at risk for dementia. It would then be possible to address in clinical trials whether effective treatment of the depression can delay the onset of dementia.


Subject(s)
Dementia/immunology , Depression/immunology , Dementia/pathology , Depression/pathology , Humans , Inflammation/immunology , Inflammation/pathology
18.
Mol Psychiatry ; 25(7): 1457-1468, 2020 07.
Article in English | MEDLINE | ID: mdl-29899546

ABSTRACT

There exists little human neuroscience research to explain why some individuals lose their appetite when they become depressed, while others eat more. Answering this question may reveal much about the various pathophysiologies underlying depression. The present study combined neuroimaging, salivary cortisol, and blood markers of inflammation and metabolism collected prior to scanning. We compared the relationships between peripheral endocrine, metabolic, and immune signaling and brain activity to food cues between depressed participants experiencing increased (N = 23) or decreased (N = 31) appetite and weight in their current depressive episode and healthy control participants (N = 42). The two depression subgroups were unmedicated and did not differ in depression severity, anxiety, anhedonia, or body mass index. Depressed participants experiencing decreased appetite had higher cortisol levels than subjects in the other two groups, and their cortisol values correlated inversely with the ventral striatal response to food cues. In contrast, depressed participants experiencing increased appetite exhibited marked immunometabolic dysregulation, with higher insulin, insulin resistance, leptin, CRP, IL-1RA, and IL-6, and lower ghrelin than subjects in other groups, and the magnitude of their insulin resistance correlated positively with the insula response to food cues. These findings provide novel evidence linking aberrations in homeostatic signaling pathways within depression subtypes to the activity of neural systems that respond to food cues and select when, what, and how much to eat. In conjunction with prior work, the present findings strongly support the existence of pathophysiologically distinct depression subtypes for which the direction of appetite change may be an easily measured behavioral marker.


Subject(s)
Appetite , Depression/immunology , Depression/metabolism , Adolescent , Adult , Appetite/immunology , C-Reactive Protein/analysis , Depression/blood , Depression/classification , Female , Ghrelin/blood , Humans , Hydrocortisone/analysis , Inflammation/blood , Inflammation/complications , Inflammation/immunology , Insulin/blood , Insulin/metabolism , Insulin Resistance , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-6/blood , Interleukin-6/immunology , Leptin/blood , Male , Middle Aged , Saliva/chemistry , Young Adult
19.
Mol Psychiatry ; 25(6): 1260-1274, 2020 06.
Article in English | MEDLINE | ID: mdl-31375779

ABSTRACT

Immune dysregulation, specifically of inflammatory processes, has been linked to behavioral symptoms of depression in both human and rodent studies. Here, we evaluated the antidepressant effects of immunization with altered peptide ligands of myelin basic protein (MBP)-MBP87-99[A91, A96], MBP87-99[A91], and MBP87-99[R91, A96]-in different models of depression and examined the mechanism by which these peptides protect against stress-induced depression. We found that a single dose of subcutaneously administered MBP87-99[A91, A96] produced antidepressant-like effects by decreasing immobility in the forced swim test and by reducing the escape latency and escape failures in the learned helplessness paradigm. Moreover, immunization with MBP87-99[A91, A96] prevented and reversed depressive-like and anxiety-like behaviors that were induced by chronic unpredictable stress (CUS). However, MBP87-99[R91, A96] tended to aggravate CUS-induced anxiety-like behavior. Chronic stress increased the production of peripheral and central proinflammatory cytokines and induced the activation of microglia in the prelimbic cortex (PrL), which was blocked by MBP87-99[A91, A96]. Immunization with MBP-derived altered peptide ligands also rescued chronic stress-induced deficits in p11, phosphorylated cyclic adenosine monophosphate response element binding protein, and brain-derived neurotrophic factor expression. Moreover, microinjections of recombinant proinflammatory cytokines and the knockdown of p11 in the PrL blunted the antidepressant-like behavioral response to MBP87-99[A91, A96]. Altogether, these findings indicate that immunization with altered MBP peptide produces prolonged antidepressant-like effects in rats, and the behavioral response is mediated by inflammatory factors (particularly interleukin-6), and p11 signaling in the PrL. Immune-neural interactions may impact central nervous system function and alter an individual's response to stress.


Subject(s)
Antidepressive Agents/chemistry , Antidepressive Agents/immunology , Depression/immunology , Depression/therapy , Immunization , Myelin Basic Protein/chemistry , Myelin Basic Protein/immunology , Animals , Antidepressive Agents/therapeutic use , Anxiety/drug therapy , Anxiety/etiology , Anxiety/immunology , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/etiology , Disease Models, Animal , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/therapeutic use , Rats , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/immunology
20.
J Psychiatry Neurosci ; 46(5): E538-E547, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34588173

ABSTRACT

BACKGROUND: Abnormalities of inflammation have been implicated in the pathophysiology of depression and suicide, based on observations of increased levels of proinflammatory cytokines in the serum of people who were depressed and died by suicide. More recently, abnormalities in cytokines and innate immunity receptors such as toll-like receptors have also been observed in the postmortem brains of people who were depressed and died by suicide. In addition to toll-like receptors, another subfamily of innate immunity receptors known as NOD-like receptors containing pyrin (NLRPs) are the most widely present NOD-like receptors in the central nervous system. NLRPs also form inflammasomes that play an important role in brain function. We studied the role of NLRPs in depression and suicide. METHODS: We determined the protein and mRNA expression of NLRP1, NLRP3 and NLRP6 and the components of their inflammasomes (i.e., adaptor molecule apoptosis-associated speck-like protein [ASC], caspase1, caspase3, interleukin [IL]-1ß and IL-18) postmortem in the prefrontal cortex of people who were depressed and died by suicide, and in healthy controls. We determined mRNA levels using quantitative polymerase chain reaction, and we determined protein expression using Western blot immunolabelling. RESULTS: We found that the protein and mRNA expression levels of NLRP1, NLRP3, NLRP6, caspase3 and ASC were significantly increased in people who were depressed and died by suicide compared to healthy controls. LIMITATIONS: Some people who were depressed and died by suicide were taking antidepressant medication at the time of their death. CONCLUSION: Similar to toll-like receptors, NLRP and its inflammasomes were upregulated in people who were depressed and died by suicide compared to healthy controls. Innate immunity receptors in general - and NLRPs and inflammasomes in particular - may play an important role in the pathophysiology of depression and suicide.


Subject(s)
Depression/immunology , Immunity, Innate , Inflammasomes , NLR Proteins/immunology , Pyrin , Suicide, Completed , Adult , Autopsy , Brain/metabolism , Cytokines/metabolism , Female , Humans , Inflammasomes/metabolism , Male , RNA, Messenger , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL