Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Biochem Biophys Res Commun ; 691: 149326, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38035406

ABSTRACT

Sleep deprivation (SD) weakens the immune system and leads to increased susceptibility to infectious or inflammatory diseases. However, it is still unclear how SD affects humoral immunity. In the present study, sleep disturbance was conducted using an sleep deprivation instrument, and the bacterial endotoxin lipopolysaccharide (LPS) was used to activate the immune response. It was found that SD-pretreatment reduced LPS-induced IgG2b+ B cells and IgG2b isotype antibody production in lymphocytes of spleen. And, SD-pretreatment decreased the proportion of CD4+T cells, production of CD4+T cells derived TGF-ß1 and its contribution in helping IgG2b production. Additionally, BMAL1 and CLOCK were selectively up-regulated in lymphocytes after SD. Importantly, BMAL1 and CLOCK deficiency contributed to TGF-ß1 expression and production of IgG2b+ B cells. Thus, our results provide a novel insight to explain the involvement of BMAL1 and CLOCK under SD stress condition, and their roles in inhibiting TGF-ß1 expression and contributing to reduction of LPS induced IgG2b production.


Subject(s)
ARNTL Transcription Factors , Antibody Formation , CLOCK Proteins , Immunoglobulin G , Sleep Deprivation , Sleep Deprivation/genetics , Sleep Deprivation/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Rats, Sprague-Dawley , Mice, Inbred C57BL , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/immunology , CLOCK Proteins/genetics , CLOCK Proteins/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Antibody Formation/drug effects , Antibody Formation/genetics , Stress, Physiological/immunology , Animals , Mice , Rats , Cells, Cultured
2.
Med Sci Sports Exerc ; 55(3): 548-557, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36563092

ABSTRACT

PURPOSE: Initial military training (IMT) is a transitionary period wherein immune function may be suppressed and infection risk heightened due to physical and psychological stress, communal living, and sleep deprivation. This study characterized changes in biomarkers of innate and adaptive immune function, and potential modulators of those changes, in military recruits during IMT. METHODS: Peripheral leukocyte distribution and mitogen-stimulated cytokine profiles were measured in fasted blood samples, Epstein-Barr (EBV), varicella zoster (VZV), and herpes simplex 1 (HSV1) DNA was measured in saliva by quantitative polymerase chain reaction as an indicator of latent herpesvirus reactivation, and diet quality was determined using the healthy eating index measured by food frequency questionnaire in 61 US Army recruits (97% male) at the beginning (PRE) and end (POST) of 22-wk IMT. RESULTS: Lymphocytes and terminally differentiated cluster of differentiation (CD)4+ and CD8+ T cells increased PRE to POST, whereas granulocytes, monocytes, effector memory CD4+ and CD8+ T cells, and central memory CD8+ T cells decreased ( P ≤ 0.02). Cytokine responses to anti-CD3/CD28 stimulation were higher POST compared with PRE, whereas cytokine responses to lipopolysaccharide stimulation were generally blunted ( P < 0.05). Prevalence of EBV reactivation was higher at POST ( P = 0.04), but neither VZV nor HSV1 reactivation was observed. Diet quality improvements were correlated with CD8+ cell maturation and blunted proinflammatory cytokine responses to anti-CD3/CD28 stimulation. CONCLUSIONS: Lymphocytosis, maturation of T-cell subsets, and increased T-cell reactivity were evident POST compared with PRE IMT. Although EBV reactivation was more prevalent at POST, no evidence of VZV or HSV1 reactivation, which are more common during severe stress, was observed. Findings suggest increases in the incidence of EBV reactivation were likely appropriately controlled by recruits and immune-competence was not compromised at the end of IMT.


Subject(s)
Military Personnel , Physical Exertion , Sleep Deprivation , Stress, Psychological , Female , Humans , Male , CD28 Antigens/blood , CD8-Positive T-Lymphocytes/metabolism , Cytokines/blood , Stress, Psychological/immunology , Sleep Deprivation/immunology , CD4-Positive T-Lymphocytes/metabolism , Physical Exertion/immunology
3.
Commun Biol ; 4(1): 1325, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824394

ABSTRACT

Poor sleep has become an important public health issue. With loss of sleep durations, poor sleep has been linked to the increased risks for diseases. Here we employed mass cytometry and single-cell RNA sequencing to obtain a comprehensive human immune cells landscape in the context of poor sleep, which was analyzed in the context of subset composition, gene signatures, enriched pathways, transcriptional regulatory networks, and intercellular interactions. Participants subjected to staying up had increased T and plasma cell frequency, along with upregulated autoimmune-related markers and pathways in CD4+ T and B cells. Additionally, staying up reduced the differentiation and immune activity of cytotoxic cells, indicative of a predisposition to infection and tumor development. Finally, staying up influenced myeloid subsets distribution and induced inflammation development and cellular senescence. These findings could potentially give high-dimensional and advanced insights for understanding the cellular and molecular mechanisms of pathologic conditions related to poor sleep.


Subject(s)
Cellular Senescence/immunology , Inflammation/etiology , Leukocytes, Mononuclear/immunology , Sleep Deprivation/immunology , Sleep/immunology , Adult , Female , Flow Cytometry , Humans , Male , Middle Aged , Single-Cell Analysis
4.
Bioengineered ; 12(1): 7920-7928, 2021 12.
Article in English | MEDLINE | ID: mdl-34622713

ABSTRACT

We aimed to assess the effects of dexmedetomidine (DEX) on postoperative cognitive function of sleep deprivation (SD) rats based on changes in inflammatory response. Male rats were randomly divided into blank control (C), SD, DEX, and SD+DEX groups. The SD model was established through intraperitoneal injection of DEX. The escape latency was detected through Morris water maze test daily, and the mechanical withdrawal threshold and thermal withdrawal latency were detected for 8 d. The content of malondialdehyde (MDA) and activity of superoxide dismutase (SOD) in hippocampus homogenate were determined, and the morphological changes in neurons were detected through Nissl staining. The concentration of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and IL-6 in the hippocampus was detected by enzyme-linked immunosorbent assay, and the Rac1/protein kinase B (AKT)/nuclear factor-κB (NF-κB) expressions were detected by Western blotting. The changes in immunofluorescence localization of NF-κB were observed by confocal microscopy. Compared with SD group, the escape latency was shortened, original platform-crossing times increased, MDA content declined, SOD activity rose, neurons were arranged orderly and number of Nissl bodies increased in the hippocampal CA1 region, levels of IL-1ß, TNF-α, and IL-6 in the hippocampus decreased, Rac1/AKT/NF-κB expressions were down-regulated, and proportion of NF-κB entering the nucleus declined in SD+DEX group (P < 0.05). DEX can effectively alleviate postoperative hippocampal inflammation and improve cognitive function of SD rats. The ability of DEX to relieve oxidative stress of hippocampal neurons, restore damaged cells, and reduce hippocampal inflammation in SD rats may be related to the Rac1/AKT/NF-κB pathway.


Subject(s)
Dexmedetomidine/administration & dosage , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Postoperative Complications/drug therapy , Sleep Deprivation/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Cognition/drug effects , Dexmedetomidine/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Hippocampus/immunology , Injections, Intraperitoneal , Male , Malondialdehyde/metabolism , Morris Water Maze Test/drug effects , NF-kappa B/metabolism , Neuropeptides/metabolism , Oxidative Stress/drug effects , Postoperative Complications/immunology , Postoperative Complications/psychology , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Sleep Deprivation/genetics , Sleep Deprivation/immunology , Sleep Deprivation/psychology , Superoxide Dismutase/metabolism , rac1 GTP-Binding Protein/metabolism
5.
Front Immunol ; 12: 727959, 2021.
Article in English | MEDLINE | ID: mdl-34539666

ABSTRACT

Sleep disturbance is common in patients with cancer and is associated with poor prognosis. However, the effects of sleep deprivation (SD) on immune surveillance during the development of hepatocellular carcinoma (HC) and the underlying mechanisms are not known. This was investigated in the present study using mouse models of SD and tumorigenesis. We determined that acute and chronic sleep deprivation (CSD) altered the relative proportions of various immune cell types in blood and peripheral organs. CSD increased tumor volume and weight, an effect that was enhanced with increasing CSD time. Expression of the cell proliferation marker Ki-67 was elevated in tumor tissues, and tumor cell infiltration into adjacent muscles was enhanced by CSD. Multicolor flow cytometry analysis revealed that CSD significantly reduced the numbers of antitumor CD3+ T cells and natural killer (NK) cells and increased that of immunosuppressive CD11b+ cells infiltrating into the tumor microenvironment from the spleen via the peripheral blood. These results indicate that CSD impairs immune surveillance mechanisms and promotes immunosuppression in the tumor microenvironment to accelerate tumor growth, underscoring the importance of alleviating sleep disturbance in HC patients in order to prevent HC progression.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Sleep Deprivation/immunology , Tumor Escape , Tumor Microenvironment/immunology , Acute Disease , Animals , CD11b Antigen/metabolism , CD3 Complex/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chronic Disease , Disease Models, Animal , Disease Progression , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice, Inbred C57BL , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Tumor Burden
7.
Mech Ageing Dev ; 192: 111388, 2020 12.
Article in English | MEDLINE | ID: mdl-33080282

ABSTRACT

OBJECTIVE: Examine subjective sleep quality and inflammation among healthy older adults participating in the Australian Research Council Longevity Intervention (ARCLI). METHODS: Data was taken from a sub-set of 232 participants aged between 60-70 years (M = 65.88 ± SD 4.08 years) who participated in the baseline assessment phase of the Australian Research Council Longevity Intervention (ARCLI) study. Subjective sleep was assessed via the Leeds Sleep Evaluation Questionnaire (LSEQ). Inflammatory markers (TNF-α, IL-1ß, IL-6, IL-10, IL-2, IFN-γ, IL-4, hs-CRP) were derived from whole blood. Correlation and multiple regression analyses were used to examine associations between each of the four sleep outcome variables and inflammatory outcomes, examined as a group and following gender stratification. RESULTS: Difficulties getting to sleep were independently associated with higher IL-2 [F(1,156) = 4.62, adjusted R2 = 0.02, p = 0.03] and IL-1ß [F(1,141) = 8.52, adjusted R2 = 0.05, p = 0.004] (whole group). Difficulties getting to sleep were associated with greater IL-1ß [males: F(1,58) = 7.36, adjusted R2 = 0.097 p = 0.009; females: F (1,81) = 4.25, R2 = 0.038, p = 0.04], and negatively associated with hs-CRP (women) [F (1,129) = 4.71, R2 = 0.028, p = 0.032]. DISCUSSION: Subjective sleep-onset difficulties are associated with systemic inflammation.


Subject(s)
C-Reactive Protein/analysis , Diagnostic Self Evaluation , Interleukins/blood , Sleep Deprivation , Systemic Inflammatory Response Syndrome , Aged , Australia/epidemiology , Correlation of Data , Cross-Sectional Studies , Female , Humans , Male , Risk Factors , Self Report/statistics & numerical data , Sleep Deprivation/diagnosis , Sleep Deprivation/immunology , Sleep Deprivation/psychology , Sleep Hygiene/physiology , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/diagnosis
8.
Aging (Albany NY) ; 12(11): 10457-10472, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32433038

ABSTRACT

Sleep disorder has become a prevalent issue in current society and is connected with the deterioration of neurobehaviors such as mood, cognition and memory. Ellagic acid (EA) is a phenolic phytoconstituent extracted from grains and fruits that has potent neuroprotective properties. This research aimed to study the alleviative effect and mechanism of EA on memory impairment and anxiety caused by sleep deprivation (SD). EA ameliorated behavioral abnormalities in SD mice, associated with increased dendritic spine density, and reduced shrinkage and loss of hippocampal neurons. EA reduced the inflammatory response and oxidative stress injury caused by SD, which may be related to activation of the Nrf2/HO-1 pathway and mitigation of the TLR4-induced inflammatory response. In addition, EA significantly reduced the mortality and ROS levels in glutamate (Glu)-induced hippocampal neuron injury, and these effects of EA were enhanced in TLR4 siRNA-transfected neurons. However, knockdown of Nrf2 dramatically restrained the protective impact of EA on Glu-induced toxicity. Taken together, EA alleviated memory impairment and anxiety in sleep-deprived mice potentially by inhibiting TLR4 and activating Nrf2. Our findings suggested that EA may be a promising nutraceutical ingredient to prevent cognitive impairment and anxiety caused by sleep loss.


Subject(s)
Anxiety/prevention & control , Cognitive Dysfunction/prevention & control , Ellagic Acid/administration & dosage , Neuroprotective Agents/administration & dosage , Sleep Deprivation/complications , Animals , Anxiety/immunology , Anxiety/pathology , Cells, Cultured , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Dietary Supplements , Disease Models, Animal , Gene Knockdown Techniques , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Primary Cell Culture , Reactive Oxygen Species/metabolism , Sleep Deprivation/diet therapy , Sleep Deprivation/immunology , Sleep Deprivation/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
10.
Aging (Albany NY) ; 12(6): 5318-5335, 2020 03 22.
Article in English | MEDLINE | ID: mdl-32200357

ABSTRACT

Major abdominal procedures could induce dysfunction in the immune system and lead to postoperative immunosuppression. Sleep dysfunction is associated with impaired immune activity. However, the effects of postoperative sleep dysfunction on postoperative immune function remain unclear. In this study, we found that sleep-restriction (SR) after surgery increased the spleen weight and the percentage of myeloid-derived suppressor cells (MDSCs) in the spleen, and inhibited splenic CD8+ T cells activity, which was via inhibiting subdiaphragmatic vagus nerve (SVN)-mediated trefoil factor 2 (TFF2) expression in the spleen of aged mice. Dexmedetomidine could alleviate SR-induced these changes via modulating gut microbiota, which acted through SVN. Moreover, we showed essential roles of splenic TFF2 in attenuating SR-induced reduced protective ability against Escherichia coli (E. coli) pneumonia, increased expression of IL-4 and IL-13 in the lung and M2 polarization of alveolar macrophages (AMs), and decreased phagocytic activity of AMs. Dexmedetomidine improved SR-induced reduced protective ability against E. coli pneumonia via splenic TFF2, and subsequently decreasing IL-4 and IL-13 expression in the lung via modulating gut microbiota/SVN, increasing the compromised phagocytic activity of AMs, and ultimately decreasing M2 polarization of AMs. Taken together, dexmedetomidine-induced increase in splenic TFF2 expresssion could alleviate SR-induced exaggeration of postoperative immunosuppression.


Subject(s)
Dexmedetomidine/pharmacology , Immune Tolerance/drug effects , Postoperative Complications/immunology , Sleep Deprivation/immunology , Trefoil Factor-2/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Escherichia coli/metabolism , Gastrointestinal Microbiome/immunology , Hypnotics and Sedatives/pharmacology , Lymphocyte Activation , Macrophages , Macrophages, Alveolar , Mice , Sleep , Spleen/immunology
11.
Int Immunopharmacol ; 78: 106041, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31835081

ABSTRACT

BACKGROUND: Previous research demonstrated that sleep deprivation (SD) resulted in intestinal homeostasis disorder in colon. The present study was further performed to clarify the role of melatonin in SD-induced small intestinal (SI) mucosal injury. METHODS: We successfully established a multiplatform 72 h SD mouse model with or without melatonin supplementation to explore the improvement of melatonin in the destruction of SI induced by SD. RESULTS: Melatonin supplementation suppressed an increase of corticosterone level and a decrease of melatonin level caused by SD. Meanwhile, we observed that melatonin supplementation in sleep deprived mice markedly reversed a decrease of the villi length/crypt depth (V/C) ratio and the number of goblet cells, PCNA positive cells, the expressions of MUC2 and tight junction proteins, as well as an upregulation of the expressions of autophagic proteins in the duodenum, jejunum and ileum. Furthermore, melatonin supplementation inverted the SD-induced the decline of antioxidant enzyme activities (T-AOC and CAT etc) and anti-inflammatory cytokines (IL-10 and IFN-γ) and the increase of oxidative product MDA, pro-inflammatory cytokines (IL-6 and TNF-α), p-P65 and p-IκB proteins in the SI. CONCLUSIONS: These findings suggested that melatonin may be used as a probiotic agent to reverse SD-induced SI mucosa injury by suppressing oxidative stress and NF-κB pathway activation.


Subject(s)
Enteritis/prevention & control , Melatonin/administration & dosage , Probiotics/administration & dosage , Sleep Deprivation/drug therapy , Animals , Disease Models, Animal , Enteritis/immunology , Enteritis/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/immunology , Intestine, Small/pathology , Male , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Sleep Deprivation/complications , Sleep Deprivation/immunology
12.
Brain Behav Immun ; 82: 129-144, 2019 11.
Article in English | MEDLINE | ID: mdl-31408672

ABSTRACT

BACKGROUND: An association between neuroinflammation, reduced adult neurogenesis, and cognitive impairment has been established in sleep deprivation (SD). Complement receptors are expressed on neuronal and glial cells, thus, regulate the neuroinflammation, neurogenesis and learning/memory. However, understanding of the effect of SD on the brain-immune system interaction associated with cognitive dysfunction and its mechanisms is obscure. We hypothesized that complement activation induced changes in inflammatory and neurogenesis related proteins might be involved in the cognitive impairment during SD. METHODOLOGY: Adult male Sprague Dawley rats were used. Rats were sleep deprived for 48 h using a novel automated SD apparatus. Dosage of BrdU (50 mg/kg/day, i.p. in 0.07 N NaOH), complement C3a receptor antagonist (C3aRA; SB290157; 1 mg/kg/day, i.p.) in 1.16% v/v PBS and complement C5a receptor antagonist (C5aRA; W-54011; 1 mg/kg/day, i.p.) in normal saline were used. Rats were subjected to spatial memory evaluation following SD. Hippocampal tissue was collected for biochemical, molecular, and immunohistochemical studies. T-test and ANOVA were used for the statistical analysis. RESULTS: An up-regulation in the levels of complement components (C3, C5, C3a, C5a) and receptors (C3aR and C5aR) in hippocampus, displayed the complement activation during SD. Selective antagonism of C3aR/C5aR improved the spatial memory performance of sleep-deprived rats. C3aR antagonist (C3aRA) or C5aR antagonist (C5aRA) treatment inhibited the gliosis, maintained inflammatory cytokines balance in hippocampus during SD. Complement C3aR/C5aR antagonism improved hippocampal adult neurogenesis via up-regulating the BDNF level following SD. Administration of C3aRA and C5aRA significantly maintained synaptic homeostasis in hippocampus after SD. Gene expression analysis showed down-regulation in the mRNA levels of signal transduction pathways (Notch and Wnt), differentiation and axogenous proteins, which were found to be improved after C3aRA/C5aRA treatment. These findings were validated at protein and cellular level. Changes in the corticosterone level and ATP-adenosine-NO pathway were established as the key mechanisms underlying complement activation mediated consequences of SD. CONCLUSION: Our study suggests complement (C3a-C3aR and C5a-C5aR) activation as the novel mechanism underlying spatial memory impairment via promoting neuroinflammation and adult neurogenesis decline in hippocampus during SD, thereby, complement (C3aR/C5aR) antagonist may serve as the novel therapeutics to improve the SD mediated consequences.


Subject(s)
Complement Activation/immunology , Neuroimmunomodulation/physiology , Sleep Deprivation/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Complement Activation/physiology , Complement C3a/metabolism , Hippocampus/metabolism , Male , Neurogenesis/immunology , Neurogenesis/physiology , Neuroimmunomodulation/immunology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Complement/metabolism , Signal Transduction/physiology , Sleep Deprivation/immunology , Spatial Memory/physiology , Temporal Lobe/metabolism
13.
J Allergy Clin Immunol ; 144(6): 1584-1594.e2, 2019 12.
Article in English | MEDLINE | ID: mdl-31319102

ABSTRACT

BACKGROUND: Peanut allergy causes severe and fatal reactions. Current food allergen labeling does not address these risks adequately against the burden of restricting food choice for allergic patients because of limited data on thresholds of reactivity and the influence of everyday factors. OBJECTIVE: We estimated peanut threshold doses for a United Kingdom population with peanut allergy and examined the effect of sleep deprivation and exercise. METHODS: In a crossover study, after blind challenge, participants with peanut allergy underwent 3 open peanut challenges in random order: with exercise after each dose, with sleep deprivation preceding challenge, and with no intervention. Primary outcome was the threshold dose triggering symptoms (in milligrams of protein). Primary analysis estimated the difference between the nonintervention challenge and each intervention in log threshold (as percentage change). Dose distributions were modeled, deriving eliciting doses in the population with peanut allergy. RESULTS: Baseline challenges were performed in 126 participants, 100 were randomized, and 81 (mean age, 25 years) completed at least 1 further challenge. The mean threshold was 214 mg (SD, 330 mg) for nonintervention challenges, and this was reduced by 45% (95% CI, 21% to 61%; P = .001) and 45% (95% CI, 22% to 62%; P = .001) for exercise and sleep deprivation, respectively. Mean estimated eliciting doses for 1% of the population were 1.5 mg (95% CI, 0.8-2.5 mg) during nonintervention challenge (n = 81), 0.5 mg (95% CI, 0.2-0.8 mg) after sleep, and 0.3 mg (95% CI, 0.1-0.6 mg) after exercise. CONCLUSION: Exercise and sleep deprivation each significantly reduce the threshold of reactivity in patients with peanut allergy, putting them at greater risk of a reaction. Adjusting reference doses using these data will improve allergen risk management and labeling to optimize protection of consumers with peanut allergy.


Subject(s)
Exercise , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/physiopathology , Sleep Deprivation/immunology , Adolescent , Adult , Cross-Over Studies , Female , Humans , Male , Middle Aged , Peanut Hypersensitivity/pathology , Sleep Deprivation/pathology , United Kingdom
14.
Sleep Breath ; 23(4): 1331-1339, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31041780

ABSTRACT

PURPOSE: The state of knowledge about the effect of sleep deprivation on the immune system is scarce and conflicting. It would therefore be useful to investigate the consequences of sleep deprivation on the immune system. We have studied the effect of sleep deprivation on the changes in neutrophil functions, and the ex vivo proliferative pattern of CD4+ T lymphocytes in relationship with blood cytokine and chemokine levels due to the crucial role of these cells in mounting potent immune responses. METHODS: Healthy volunteers were followed for 3 weeks. They had normal sleep in weeks 1 and 3 and they were sleep-deprived on week 2, sleeping < 6 h per 24 h, a pattern similar to sleep behaviors of many chronically sleep-deprived individuals. We assessed the levels of Th1/Th2 and inflammatory cytokines and chemokines, CD4+ T cells, and the NADPH oxidase activation and phagocytic functions in neutrophils. RESULTS: Our results suggest that sleep deprivation leads to a decreased neutrophil capacity to phagocytose bacteria and activate NADPH oxidase (p < 0.05). Sleep deprivation was associated with a potential increase in CXCL9 levels and decrease in CXCL10/CXCL9 and CCL5/CXCL9 ratios (p < 0.05). Furthermore, our results suggest that the decrease in CD4+ T cell due to sleep deprivation was not associated with changes in their proliferation as observed by Ki67 levels, but rather, it correlated with changes in CXCL10/CXCL9 ratio (p < 0.05). CONCLUSIONS: Sleep deprivation may lead to a decreased phagocytosis and NADPH oxidase activity in neutrophils and a decrease in the levels of CD4+ T cells which is related to changes in the Th1-related chemokine balance.


Subject(s)
CD4 Lymphocyte Count , Chemokines/physiology , Neutrophils/physiology , Sleep Deprivation/immunology , Th1-Th2 Balance/physiology , Adult , Cell Proliferation , Cytokines/blood , Female , Humans , Male , NADPH Oxidases/blood , Phagocytosis/immunology , Reference Values
15.
Biochem Biophys Res Commun ; 513(2): 452-459, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30967262

ABSTRACT

Sleep deprivation affects the pathophysiology of immune-inflammatory skin diseases such as psoriasis. Increasing efforts are directed toward exploring potential mechanisms involving sleep deprivation and immune responses. However, studies focusing on the effects of different timing of sleep deprivation on skin inflammation are lacking. This study thus investigated the immunomodulatory effects of paradoxical sleep (PS) deprivation at different timing of psoriasiform process on skin inflammation in the psoriasis-like mouse model. Male adult C57BL/6 mice were divided into 5 groups (each n = 5): control group (CON), IMQ-S2 group [treating with 31.25 mg imiquimod (IMQ) per day for 5 days with 48 h of PS deprivation on the fourth and fifth day], S2-IMQ group (treating with 31.25 mg IMQ per day for 5 days with 48 h of PS deprivation on the first and second day), 31.25 group (treating with 31.25 mg IMQ per day for 5 days) and 62.5 group (treating with 62.5 mg IMQ per day for 5 days). Compared with IMQ-S2 group and 31.25 group, S2-IMQ group mice had significant increase of IL-17A mRNA level in skin lesions and lymph nodes, and more severe cutaneous inflammation. However, IMQ-S2 group had the highest IL-1ß mRNA level in skin lesions and the highest IL-6 mRNA level in lymph nodes among the three groups. Results of flow cytometry showed that frequencies of γδT cell, IL-17A+γδT cell, dendritic cell (DC) and MHCⅡ+ DC in lymph nodes of S2-IMQ group were significantly higher than IMQ-S2 group and 31.25 group, so was the frequency of γδT cells in skin lesions. However, the frequency of DCs in skin lesions of S2-IMQ group was significantly lower than IMQ-S2 group. These data suggest that PS deprivation at the early stage rather than the late stage of psoriasiform process exacerbates the skin inflammation through modulation of the immune system, which may involve that IL-1ß and IL-6 stimulated by PS deprivation in turn increasing IL-17 expression through activation and proliferation of γδT cells and DCs migration.


Subject(s)
Inflammation/complications , Psoriasis/complications , Skin/pathology , Sleep Deprivation/complications , Animals , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Inflammation/immunology , Inflammation/pathology , Interleukin-17/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Male , Mice, Inbred C57BL , Psoriasis/immunology , Psoriasis/pathology , Skin/immunology , Sleep Deprivation/immunology , Sleep Deprivation/pathology
16.
J Pineal Res ; 67(1): e12574, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30929267

ABSTRACT

Intestinal diseases caused by sleep deprivation (SD) are severe public health threats worldwide. This study focuses on the effect of melatonin on intestinal mucosal injury and microbiota dysbiosis in sleep-deprived mice. Mice subjected to SD had significantly elevated norepinephrine levels and decreased melatonin content in plasma. Consistent with the decrease in melatonin levels, we observed a decrease of antioxidant ability, down-regulation of anti-inflammatory cytokines and up-regulation of pro-inflammatory cytokines in sleep-deprived mice, which resulted in colonic mucosal injury, including a reduced number of goblet cells, proliferating cell nuclear antigen-positive cells, expression of MUC2 and tight junction proteins and elevated expression of ATG5, Beclin1, p-P65 and p-IκB. High-throughput pyrosequencing of 16S rRNA demonstrated that the diversity and richness of the colonic microbiota were decreased in sleep-deprived mice, especially in probiotics, including Akkermansia, Bacteroides and Faecalibacterium. However, the pathogen Aeromonas was markedly increased. By contrast, supplementation with 20 and 40 mg/kg melatonin reversed these SD-induced changes and improved the mucosal injury and dysbiosis of the microbiota in the colon. Our results suggest that the effect of SD on intestinal barrier dysfunction might be an outcome of melatonin suppression rather than a loss of sleep per se. SD-induced intestinal barrier dysfunction involved the suppression of melatonin production and activation of the NF-κB pathway by oxidative stress.


Subject(s)
Colon , Gastrointestinal Microbiome/immunology , Intestinal Diseases , Intestinal Mucosa , Melatonin/immunology , Sleep Deprivation , Animals , Colon/immunology , Colon/microbiology , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Probiotics , Sleep Deprivation/immunology , Sleep Deprivation/microbiology
17.
Physiol Rev ; 99(3): 1325-1380, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30920354

ABSTRACT

Sleep and immunity are bidirectionally linked. Immune system activation alters sleep, and sleep in turn affects the innate and adaptive arm of our body's defense system. Stimulation of the immune system by microbial challenges triggers an inflammatory response, which, depending on its magnitude and time course, can induce an increase in sleep duration and intensity, but also a disruption of sleep. Enhancement of sleep during an infection is assumed to feedback to the immune system to promote host defense. Indeed, sleep affects various immune parameters, is associated with a reduced infection risk, and can improve infection outcome and vaccination responses. The induction of a hormonal constellation that supports immune functions is one likely mechanism underlying the immune-supporting effects of sleep. In the absence of an infectious challenge, sleep appears to promote inflammatory homeostasis through effects on several inflammatory mediators, such as cytokines. This notion is supported by findings that prolonged sleep deficiency (e.g., short sleep duration, sleep disturbance) can lead to chronic, systemic low-grade inflammation and is associated with various diseases that have an inflammatory component, like diabetes, atherosclerosis, and neurodegeneration. Here, we review available data on this regulatory sleep-immune crosstalk, point out methodological challenges, and suggest questions open for future research.


Subject(s)
Immune System/physiology , Immunity/physiology , Sleep/immunology , Sleep/physiology , Animals , Homeostasis , Humans , Immunity, Innate/physiology , Sleep Deprivation/immunology
18.
Brain Res ; 1714: 81-87, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30677408

ABSTRACT

Sleep deprivation impairs hippocampal neurogenesis, but the underlying mechanisms are inadequately understood. Sleep deprivation is associated with an increased production of pro-inflammatory factors. In this study, we demonstrate that acute rapid eye movement (REM) sleep deprivation in mice for 3 days leads to increased expression of interleukin (IL)-17A, IL-17F and activation of p38 MAPK pathway in the hippocampus, together with suppressed cell proliferation in the dentate gyrus. Similarly, activation of p38 MAPK and suppressed cell proliferation in the dentate gyrus were observed after administration of recombinant IL-17 in mice without sleep deprivation. Pharmacological blockade of the p38 MAPK after sleep deprivation mitigates sleep deprivation-induced ablation of cell proliferation in the dentate gyrus with unaltered expression of IL-17A and IL-17F. In addition, hippocampal neural progenitor cells express IL-17 receptor A (IL-17RA) and IL-17 receptor C (IL-17RC). These findings suggest that acute REM sleep deprivation suppresses proliferation of adult hippocampal neural progenitor cells by a mechanism involving IL-17 and p38 MAPK signaling.


Subject(s)
Interleukin-17/metabolism , Neural Stem Cells/metabolism , Sleep Deprivation/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adult Stem Cells/metabolism , Animals , Cell Proliferation/physiology , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Hippocampus/metabolism , Hippocampus/physiology , Imidazoles/pharmacology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/immunology , Neurogenesis/physiology , Pyridines/pharmacology , Receptors, Interleukin-17/metabolism , Sleep Deprivation/immunology
19.
Neuroimmunomodulation ; 25(2): 59-67, 2018.
Article in English | MEDLINE | ID: mdl-30007965

ABSTRACT

OBJECTIVES: Sleep regulates immune function reciprocally and can affect the parameters that are directly involved in the immune response. Sleep deprivation is considered to be a stress-causing factor and is associated with impaired immune activity. It causes increased glucocorticoid concentrations by activating the hypothalamic-pituitary-adrenal axis; this can lead to a series of disorders that are associated with the prolonged or increased secretion of these hormones. The aim of this study was to evaluate the effects of sleep restriction (SR) on the development of pulmonary experimental metastasis and the modulation of the tumor immune response. METHODS: The SR protocol was accomplished by depriving C57BL/6 male mice of sleep for 18 h/day for 2, 7, 14, and 21 days. The modified multiple-platforms method was used for SR. RESULTS: The results showed that cytotoxic cells (i.e., natural killer [NK] and CD8+ T cells) were reduced in number and regulatory T cells were predominant in the tumor microenvironment. Sleep-restricted mice also exhibited a reduced number of dendritic cells in their lymph nodes, which may have contributed to the ineffective activation of tumor-specific T cells. Peripheral CD4+ and CD8+ T cells were also reduced in the sleep-restricted mice, thus indicating an immunosuppressive status. CONCLUSIONS: Sleep dep-rivation induces failure in the activity of cells that are im-portant to the tumor immune response, both in the tumor microenvironment and on the periphery. This leads to the early onset and increased growth rate of lung metastasis.


Subject(s)
Immunity, Cellular/immunology , Lung Neoplasms/immunology , Lymphocytes/immunology , Sleep Deprivation/immunology , Tumor Microenvironment/immunology , Animals , Lung Neoplasms/pathology , Lymphocytes/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Sleep Deprivation/pathology
20.
J Neuroimmunol ; 316: 98-106, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29406850

ABSTRACT

BACKGROUND: Postpartum is a period of unique psychosocial stress characterized by sleep disturbance, risk for depressed mood, and heightened parenting stress. However, data on effects of these exposures on inflammatory immune function are limited. METHODS: This study examined associations among sleep, psychosocial stress (i.e., parenting stress, general perceived stress), mood (i.e., depressive symptoms), serum cytokine levels, and LPS-stimulated proinflammatory cytokine production among 69 women (32 African American, 37 White) assessed at 7-10weeks postpartum. RESULTS: No associations between behavioral measures and serum cytokine levels were observed among women of either race. In African American women, but not Whites, poorer sleep quality, greater parenting stress, and greater depressive symptoms were associated with greater LPS-stimulated IL-6 and IL-8 production (ps≤0.05). Also in African Americans, greater general perceived stress was associated with greater IL-8 production, and greater depressive symptoms with greater stimulated TNF-α production (ps≤0.05). Simple mediation models highlighted the bidirectional relationship between stress and sleep in relation to inflammation among African American women. CONCLUSIONS: Significant effects of both stress/distress and poor sleep quality on proinflammatory cytokine production during postpartum were observed uniquely among African American women. These data are consistent with an allostatic load model which predicts that conditions of chronic stress impart vulnerability to dysregulated responses to novel stressor exposures. The bidirectional nature of the stress-sleep relationship has clinical relevance. Studies examining whether interventions focused on one or both of these psychological factors during postpartum is beneficial for inflammatory profiles would be informative. In addition, examination of these models in relation to maternal health at postpartum, including delivery related wounds and other infections, is warranted.


Subject(s)
Cytokines/blood , Postpartum Period/immunology , Postpartum Period/psychology , Sleep Deprivation/immunology , Stress, Psychological/immunology , Black or African American , Female , Humans , Lipopolysaccharides/pharmacology , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...