1
|
Guo DZ, Chen Y, Meng Y, Bian JJ, Wang Y, Wang JF. Bidirectional Interaction of Sepsis and Sleep Disorders: The Underlying Mechanisms and Clinical Implications. Nat Sci Sleep 2024; 16:1665-1678. [PMID: 39444661 PMCID: PMC11498039 DOI: 10.2147/nss.s485920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024] Open
Abstract
Sepsis is defined as life-threatening organ injury induced by infection, with high incidence and mortality. Sleep disorder is prevalent in septic patients and approximately 50% of patients with sepsis may develop atypical sleep patterns, but many of them may have been underdiagnosed by physicians. Sleep disorders and sepsis exhibit a close bidirectional relationship, with each condition significantly influencing the other. Conversely, sleep deprivation, sleep dysrhythmia and sleep fragmentation have been shown to impact the outcome of sepsis. This review endeavors to offer a comprehensive understanding of the intricate mechanisms that underpin the interplay between sepsis and sleep disorders, in addition to exploring potential clinical intervention strategies that could enhance outcomes for patients suffering from sepsis.
Collapse
Affiliation(s)
- De-Zhi Guo
- School of Basic Medicine, Naval Medical University, Shanghai, People’s Republic of China
| | - Yu Chen
- School of Basic Medicine, Naval Medical University, Shanghai, People’s Republic of China
| | - Yan Meng
- Department of Intensive Care, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Jin-Jun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
2
|
Hasan ZW, Nguyen VT, Ashley NT. Effect of glucocorticoid blockade on inflammatory responses to acute sleep fragmentation in male mice. PeerJ 2024; 12:e17539. [PMID: 38952964 PMCID: PMC11216221 DOI: 10.7717/peerj.17539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/19/2024] [Indexed: 07/03/2024] Open
Abstract
The association between sleep and the immune-endocrine system is well recognized, but the nature of that relationship is not well understood. Sleep fragmentation induces a pro-inflammatory response in peripheral tissues and brain, but it also activates the hypothalamic-pituitary-adrenal (HPA) axis, releasing glucocorticoids (GCs) (cortisol in humans and corticosterone in mice). It is unclear whether this rapid release of glucocorticoids acts to potentiate or dampen the inflammatory response in the short term. The purpose of this study was to determine whether blocking or suppressing glucocorticoid activity will affect the inflammatory response from acute sleep fragmentation (ASF). Male C57BL/6J mice were injected i.p. with either 0.9% NaCl (vehicle 1), metyrapone (a glucocorticoid synthesis inhibitor, dissolved in vehicle 1), 2% ethanol in polyethylene glycol (vehicle 2), or mifepristone (a glucocorticoid receptor antagonist, dissolved in vehicle 2) 10 min before the start of ASF or no sleep fragmentation (NSF). After 24 h, samples were collected from brain (prefrontal cortex, hypothalamus, hippocampus) and periphery (liver, spleen, heart, and epididymal white adipose tissue (EWAT)). Proinflammatory gene expression (TNF-α and IL-1β) was measured, followed by gene expression analysis. Metyrapone treatment affected pro-inflammatory cytokine gene expression during ASF in some peripheral tissues, but not in the brain. More specifically, metyrapone treatment suppressed IL-1β expression in EWAT during ASF, which implies a pro-inflammatory effect of GCs. However, in cardiac tissue, metyrapone treatment increased TNF-α expression in ASF mice, suggesting an anti-inflammatory effect of GCs. Mifepristone treatment yielded more significant results than metyrapone, reducing TNF-α expression in liver (only NSF mice) and cardiac tissue during ASF, indicating a pro-inflammatory role. Conversely, in the spleen of ASF-mice, mifepristone increased pro-inflammatory cytokines (TNF-α and IL-1β), demonstrating an anti-inflammatory role. Furthermore, irrespective of sleep fragmentation, mifepristone increased pro-inflammatory cytokine gene expression in heart (IL-1β), pre-frontal cortex (IL-1β), and hypothalamus (IL-1β). The results provide mixed evidence for pro- and anti-inflammatory functions of corticosterone to regulate inflammatory responses to acute sleep loss.
Collapse
Affiliation(s)
- Zim Warda Hasan
- Department of Biology, Western Kentucky University, Bowling Green, KY, United States of America
| | - Van Thuan Nguyen
- Department of Biology, Western Kentucky University, Bowling Green, KY, United States of America
| | - Noah T. Ashley
- Department of Biology, Western Kentucky University, Bowling Green, KY, United States of America
| |
Collapse
|
3
|
Lau RI, Su Q, Ching JYL, Lui RN, Chan TT, Wong MTL, Lau LHS, Wing YK, Chan RNY, Kwok HYH, Ho AHY, Tse YK, Cheung CP, Li MKT, Siu WY, Liu C, Lu W, Wang Y, Chiu EOL, Cheong PK, Chan FKL, Ng SC. Fecal Microbiota Transplantation for Sleep Disturbance in Post-acute COVID-19 Syndrome. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00542-1. [PMID: 38908733 DOI: 10.1016/j.cgh.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND & AIMS Post-acute COVID-19 syndrome (PACS) is associated with sleep disturbance, but treatment options are limited. The etiology of PACS may be secondary to alterations in the gut microbiome. Here, we report the efficacy of fecal microbiota transplantation (FMT) in alleviating post-COVID insomnia symptoms in a nonrandomized, open-label prospective interventional study. METHODS Between September 22, 2022, and May 22, 2023, we recruited 60 PACS patients with insomnia defined as Insomnia Severity Index (ISI) ≥8 and assigned them to the FMT group (FMT at weeks 0, 2, 4, and 8; n = 30) or the control group (n = 30). The primary outcome was clinical remission defined by an ISI of <8 at 12 weeks. Secondary outcomes included changes in the Pittsburgh Sleep Quality Index, Generalized Anxiety Disorder-7 scale, Epworth Sleepiness Scale, Multidimensional Fatigue Inventory, blood cortisol and melatonin, and gut microbiome analysis on metagenomic sequencing. RESULTS At week 12, more patients in the FMT than the control group had insomnia remission (37.9% vs 10.0%; P = .018). The FMT group showed a decrease in ISI score (P < .0001), Pittsburgh Sleep Quality Index (P < .0001), Generalized Anxiety Disorder-7 scale (P = .0019), Epworth Sleepiness Scale (P = .0057), and blood cortisol concentration (P = .035) from baseline to week 12, but there was no significant change in the control group. There was enrichment of bacteria such as Gemmiger formicilis and depletion of microbial pathways producing menaquinol derivatives after FMT. The gut microbiome profile resembled that of the donor in FMT responders but not in nonresponders at week 12. There was no serious adverse event. CONCLUSIONS This pilot study showed that FMT could be effective and safe in alleviating post-COVID insomnia, and further clinical trials are warranted. CLINICALTRIALS gov, Number: NCT05556733.
Collapse
Affiliation(s)
- Raphaela I Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Qi Su
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Jessica Y L Ching
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Rashid N Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ting Ting Chan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Marc T L Wong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Louis H S Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yun Kwok Wing
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rachel N Y Chan
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hanson Y H Kwok
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Agnes H Y Ho
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yee Kit Tse
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Moses K T Li
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Wan Ying Siu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Chengyu Liu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Wenqi Lu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Yun Wang
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Emily O L Chiu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pui Kuan Cheong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Francis K L Chan
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Siew C Ng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Shaikh H, Ionita R, Khan U, Park Y, Jubran A, Tobin MJ, Laghi F. Effect of Atypical Sleep EEG Patterns on Weaning From Prolonged Mechanical Ventilation. Chest 2024; 165:1111-1119. [PMID: 38211699 PMCID: PMC11214907 DOI: 10.1016/j.chest.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Approximately one-third of acute ICU patients display atypical sleep patterns that cannot be interpreted by using standard EEG criteria for sleep. Atypical sleep patterns have been associated with poor weaning outcomes in acute ICUs. RESEARCH QUESTION Do patients being weaned from prolonged mechanical ventilation experience atypical sleep EEG patterns, and are these patterns linked with weaning outcomes? STUDY DESIGN AND METHODS EEG power spectral analysis during wakefulness and overnight polysomnogram were performed on alert, nondelirious patients at a long-term acute care facility. RESULTS Forty-four patients had been ventilated for a median duration of 38 days at the time of the polysomnogram study. Eleven patients (25%) exhibited atypical sleep EEG. During wakefulness, relative EEG power spectral analysis revealed higher relative delta power in patients with atypical sleep than in patients with usual sleep (53% vs 41%; P < .001) and a higher slow-to-fast power ratio during wakefulness: 4.39 vs 2.17 (P < .001). Patients with atypical sleep displayed more subsyndromal delirium (36% vs 6%; P = .027) and less rapid eye movement sleep (4% vs 11% total sleep time; P < .02). Weaning failure was more common in the atypical sleep group than in the usual sleep group: 91% vs 45% (P = .013). INTERPRETATION This study provides the first evidence that patients in a long-term acute care facility being weaned from prolonged ventilation exhibit atypical sleep EEG patterns that are associated with weaning failure. Patients with atypical sleep EEG patterns had higher rates of subsyndromal delirium and slowing of the wakeful EEG, suggesting that these two findings represent a biological signal for brain dysfunction.
Collapse
Affiliation(s)
- Hameeda Shaikh
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL
| | - Ramona Ionita
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL
| | - Usman Khan
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL
| | - Youngsook Park
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL
| | - Amal Jubran
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL; RML Specialty Hospital, Hinsdale, IL
| | - Martin J Tobin
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL.
| | - Franco Laghi
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital and Loyola University of Chicago Stritch School of Medicine, Hines, IL; RML Specialty Hospital, Hinsdale, IL
| |
Collapse
|
5
|
Rendine M, Cocci P, de Vivo L, Bellesi M, Palermo FA. Effects of Chronic Sleep Restriction on Transcriptional Sirtuin 1 Signaling Regulation in Male Mice White Adipose Tissue. Curr Issues Mol Biol 2024; 46:2144-2154. [PMID: 38534754 DOI: 10.3390/cimb46030138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic sleep restriction (CSR) is a prevalent issue in modern society that is associated with several pathological states, ranging from neuropsychiatric to metabolic diseases. Despite its known impact on metabolism, the specific effects of CSR on the molecular mechanisms involved in maintaining metabolic homeostasis at the level of white adipose tissue (WAT) remain poorly understood. Therefore, this study aimed to investigate the influence of CSR on sirtuin 1 (SIRT1) and the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway in the WAT of young male mice. Both genes interact with specific targets involved in multiple metabolic processes, including adipocyte differentiation, browning, and lipid metabolism. The quantitative PCR (qPCR) results demonstrated a significant upregulation of SIRT-1 and some of its target genes associated with the transcriptional regulation of lipid homeostasis (i.e., PPARα, PPARγ, PGC-1α, and SREBF) and adipose tissue development (i.e., leptin, adiponectin) in CSR mice. On the contrary, DNA-binding transcription factors (i.e., CEBP-β and C-myc), which play a pivotal function during the adipogenesis process, were found to be down-regulated. Our results also suggest that the induction of SIRT1-dependent molecular pathways prevents weight gain. Overall, these findings offer new, valuable insights into the molecular adaptations of WAT to CSR, in order to support increased energy demand due to sleep loss.
Collapse
Affiliation(s)
- Marco Rendine
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133 Milan, Italy
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Luisa de Vivo
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Michele Bellesi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1QU, UK
| | | |
Collapse
|
6
|
Zhang YM, Zhang MY, Wei RM, Zhang JY, Zhang KX, Luo BL, Ge YJ, Kong XY, Li XY, Chen GH. Subsequent maternal sleep deprivation aggravates neurobehavioral abnormalities, inflammation, and synaptic function in adult male mice exposed to prenatal inflammation. Front Behav Neurosci 2023; 17:1226300. [PMID: 37560531 PMCID: PMC10407227 DOI: 10.3389/fnbeh.2023.1226300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023] Open
Abstract
OBJECTIVE Studies have suggested that prenatal exposure to inflammation increases the risk of neuropsychiatric disorders, including anxiety, depression, and cognitive dysfunction. Because of anatomical and hormonal alterations, pregnant women frequently experience sleep dysfunction, which can enhance the inflammatory response. The aim of this study was to explore the effects of maternal sleep deprivation on prenatal inflammation exposure-induced behavioral phenotypes in offspring and identify the associated mechanisms. METHODS Pregnant mice received an intraperitoneal injection of lipopolysaccharide (LPS) on gestational day 15 and were subsequently subjected to sleep deprivation during gestational days 15-21. Anxiety-like behavior was evaluated by the open field test and the elevated plus maze test. Depression-like behavior was assessed by the tail suspension test and the forced swimming test. Cognitive function was determined using the Morris water maze test. The levels of markers of inflammation and synaptic function were examined employing general molecular biological techniques. RESULTS The results showed that prenatal exposure to LPS resulted in anxiety- and depression-like symptoms and learning and memory deficits, and these effects were exacerbated by maternal sleep deprivation. Furthermore, maternal sleep deprivation aggravated the prenatal LPS exposure-induced increase in the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α and decrease in the levels of postsynaptic density-95 and synaptophysin in the hippocampus. DISCUSSION Collectively, these results suggested that maternal sleep deprivation exacerbates anxiety, depression, and cognitive impairment induced by prenatal LPS exposure, effects that were associated with an inflammatory response and synaptic dysfunction.
Collapse
Affiliation(s)
- Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meng-Ying Zhang
- Department of Anesthesiology, The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ru-Meng Wei
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing-Ya Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kai-Xuan Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bao-Ling Luo
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi-Jun Ge
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Yi Kong
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
Hillman DR, Carlucci M, Charchaflieh JG, Cloward TV, Gali B, Gay PC, Lyons MM, McNeill MM, Singh M, Yilmaz M, Auckley DH. Society of Anesthesia and Sleep Medicine Position Paper on Patient Sleep During Hospitalization. Anesth Analg 2023; 136:814-824. [PMID: 36745563 DOI: 10.1213/ane.0000000000006395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This article addresses the issue of patient sleep during hospitalization, which the Society of Anesthesia and Sleep Medicine believes merits wider consideration by health authorities than it has received to date. Adequate sleep is fundamental to health and well-being, and insufficiencies in its duration, quality, or timing have adverse effects that are acutely evident. These include cardiovascular dysfunction, impaired ventilatory function, cognitive impairment, increased pain perception, psychomotor disturbance (including increased fall risk), psychological disturbance (including anxiety and depression), metabolic dysfunction (including increased insulin resistance and catabolic propensity), and immune dysfunction and proinflammatory effects (increasing infection risk and pain generation). All these changes negatively impact health status and are counterproductive to recovery from illness and operation. Hospitalization challenges sleep in a variety of ways. These challenges include environmental factors such as noise, bright light, and overnight awakenings for observations, interventions, and transfers; physiological factors such as pain, dyspnea, bowel or urinary dysfunction, or discomfort from therapeutic devices; psychological factors such as stress and anxiety; care-related factors including medications or medication withdrawal; and preexisting sleep disorders that may not be recognized or adequately managed. Many of these challenges appear readily addressable. The key to doing so is to give sleep greater priority, with attention directed at ensuring that patients' sleep needs are recognized and met, both within the hospital and beyond. Requirements include staff education, creation of protocols to enhance the prospect of sleep needs being addressed, and improvement in hospital design to mitigate environmental disturbances. Hospitals and health care providers have a duty to provide, to the greatest extent possible, appropriate preconditions for healing. Accumulating evidence suggests that these preconditions include adequate patient sleep duration and quality. The Society of Anesthesia and Sleep Medicine calls for systematic changes in the approach of hospital leadership and staff to this issue. Measures required include incorporation of optimization of patient sleep into the objectives of perioperative and general patient care guidelines. These steps should be complemented by further research into the impact of hospitalization on sleep, the effects of poor sleep on health outcomes after hospitalization, and assessment of interventions to improve it.
Collapse
Affiliation(s)
- David R Hillman
- From the West Australian Sleep Disorders Research Institute, Centre for Sleep Science, University of Western Australia, Perth, Western Australia, Australia
| | - Melissa Carlucci
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jean G Charchaflieh
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Tom V Cloward
- Division of Sleep Medicine, Intermountain Health Care and Division of Pulmonary, Critical Care and Sleep Medicine, University of Utah, Salt Lake City, Utah
| | - Bhargavi Gali
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Peter C Gay
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mayo Clinic, Rochester, Minnesota
| | - M Melanie Lyons
- Division of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | | | - Mandeep Singh
- Department of Anesthesia, Women's College Hospital, and Toronto Western Hospital, University Health Network; University of Toronto, Toronto, Ontario, Canada
| | - Meltem Yilmaz
- Department of Anesthesiology, Northwestern University, Chicago, Illinois
| | - Dennis H Auckley
- Division of Pulmonary, Critical Care and Sleep Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
8
|
Stenger S, Grasshoff H, Hundt JE, Lange T. Potential effects of shift work on skin autoimmune diseases. Front Immunol 2023; 13:1000951. [PMID: 36865523 PMCID: PMC9972893 DOI: 10.3389/fimmu.2022.1000951] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/29/2022] [Indexed: 02/16/2023] Open
Abstract
Shift work is associated with systemic chronic inflammation, impaired host and tumor defense and dysregulated immune responses to harmless antigens such as allergens or auto-antigens. Thus, shift workers are at higher risk to develop a systemic autoimmune disease and circadian disruption with sleep impairment seem to be the key underlying mechanisms. Presumably, disturbances of the sleep-wake cycle also drive skin-specific autoimmune diseases, but epidemiological and experimental evidence so far is scarce. This review summarizes the effects of shift work, circadian misalignment, poor sleep, and the effect of potential hormonal mediators such as stress mediators or melatonin on skin barrier functions and on innate and adaptive skin immunity. Human studies as well as animal models were considered. We will also address advantages and potential pitfalls in animal models of shift work, and possible confounders that could drive skin autoimmune diseases in shift workers such as adverse lifestyle habits and psychosocial influences. Finally, we will outline feasible countermeasures that may reduce the risk of systemic and skin autoimmunity in shift workers, as well as treatment options and highlight outstanding questions that should be addressed in future studies.
Collapse
Affiliation(s)
- Sarah Stenger
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Jennifer Elisabeth Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
- Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
9
|
Complexification of In Vitro Models of Intestinal Barriers, A True Challenge for a More Accurate Alternative Approach. Int J Mol Sci 2023; 24:ijms24043595. [PMID: 36835003 PMCID: PMC9958734 DOI: 10.3390/ijms24043595] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
The use of cell models is common to mimic cellular and molecular events in interaction with their environment. In the case of the gut, the existing models are of particular interest to evaluate food, toxicants, or drug effects on the mucosa. To have the most accurate model, cell diversity and the complexity of the interactions must be considered. Existing models range from single-cell cultures of absorptive cells to more complex combinations of two or more cell types. This work describes the existing solutions and the challenges that remain to be solved.
Collapse
|
10
|
Karl JP, Whitney CC, Wilson MA, Fagnant HS, Radcliffe PN, Chakraborty N, Campbell R, Hoke A, Gautam A, Hammamieh R, Smith TJ. Severe, short-term sleep restriction reduces gut microbiota community richness but does not alter intestinal permeability in healthy young men. Sci Rep 2023; 13:213. [PMID: 36604516 PMCID: PMC9816096 DOI: 10.1038/s41598-023-27463-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Sleep restriction alters gut microbiota composition and intestinal barrier function in rodents, but whether similar effects occur in humans is unclear. This study aimed to determine the effects of severe, short-term sleep restriction on gut microbiota composition and intestinal permeability in healthy adults. Fecal microbiota composition, measured by 16S rRNA sequencing, and intestinal permeability were measured in 19 healthy men (mean ± SD; BMI 24.4 ± 2.3 kg/m2, 20 ± 2 years) undergoing three consecutive nights of adequate sleep (AS; 7-9 h sleep/night) and restricted sleep (SR; 2 h sleep/night) in random order with controlled diet and physical activity. α-diversity measured by amplicon sequencing variant (ASV) richness was 21% lower during SR compared to AS (P = 0.03), but α-diversity measured by Shannon and Simpson indexes did not differ between conditions. Relative abundance of a single ASV within the family Ruminococcaceae was the only differentially abundant taxon (q = 0.20). No between-condition differences in intestinal permeability or β-diversity were observed. Findings indicated that severe, short-term sleep restriction reduced richness of the gut microbiota but otherwise minimally impacted community composition and did not affect intestinal permeability in healthy young men.
Collapse
Affiliation(s)
- J. Philip Karl
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA
| | - Claire C. Whitney
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA
| | - Marques A. Wilson
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA
| | - Heather S. Fagnant
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA
| | - Patrick N. Radcliffe
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA ,grid.410547.30000 0001 1013 9784Oak Ridge Institute of Science and Education, Oak Ridge, TN USA
| | - Nabarun Chakraborty
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Ross Campbell
- grid.507680.c0000 0001 2230 3166Geneva Foundation, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Allison Hoke
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Aarti Gautam
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Rasha Hammamieh
- grid.507680.c0000 0001 2230 3166Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD USA
| | - Tracey J. Smith
- grid.420094.b0000 0000 9341 8465Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA USA
| |
Collapse
|
11
|
Akkaoui MA, Palagini L, Geoffroy PA. Sleep Immune Cross Talk and Insomnia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:263-273. [PMID: 36949314 DOI: 10.1007/978-981-19-7376-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Sleep and immunity have bidirectional relationships. In this chapter, we review the links between sleep and immunity, focusing on immune changes occurring in the insomnia disorder. During physiological sleep, there is a decrease of pro-inflammatory cytokines (IL-1, IL-6 and TNF-α) and a decrease of anti-inflammatory cytokines (IL-4, IL-10). Examinations of ratios of pro-inflammatory and anti-inflammatory cytokines allow to identify rather a pro-inflammatory activity at the beginning of the night and confirm then anti-inflammatory during the second part of the night. Immune cells, as NK-cells, decrease in the blood, due to their migration to secondary lymphoid organs, but their activity increases. Inversely, a short sleep duration appears associated with increased inflammatory processes and increased risk of infection.Only few studies have investigated changes in immunity in patients with insomnia disorder. These studies suggest that insomnia disorder is related to deregulation of the immune system, with an increase in the level of pro-inflammatory cytokines and change in rate of secretion and a decrease in the level of lymphocyte. Insomnia treatments, particularly cognitive behavioral therapy (CBT-I), seems to have a restorative effect not only on sleep, but also on the associated inflammation. Melatonin also seems to reduce inflammation in patients suffering from insomnia disorder.More studies are necessary to better understand the pathophysiology of changes in immune system in patients suffering from insomnia disorders and their clinical implications.
Collapse
Affiliation(s)
- Marine Ambar Akkaoui
- Centre Psychiatrique d'Orientation et d'Accueil (CPOA), GHU Paris - Psychiatry & Neurosciences, Paris, France
- Etablissement Publique de Santé Mentale de Ville Evrard, Neuilly Sur Marne, France
| | - Laura Palagini
- Psychiatric Clinic, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- Psychiatric Clinic Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Pierre A Geoffroy
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hôpital Bichat - Claude Bernard, Paris, France
- GHU Paris - Psychiatry & Neurosciences, Paris, France
- Université de Paris, NeuroDiderot, Inserm, Paris, France
- CNRS UPR 3212, Institute for Cellular and Integrative Neurosciences, Strasbourg, France
| |
Collapse
|
12
|
Greening L, McBride S. A Review of Equine Sleep: Implications for Equine Welfare. Front Vet Sci 2022; 9:916737. [PMID: 36061116 PMCID: PMC9428463 DOI: 10.3389/fvets.2022.916737] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep is a significant biological requirement for all living mammals due to its restorative properties and its cognitive role in memory consolidation. Sleep is ubiquitous amongst all mammals but sleep profiles differ between species dependent upon a range of biological and environmental factors. Given the functional importance of sleep, it is important to understand these differences in order to ensure good physical and psychological wellbeing for domesticated animals. This review focuses specifically on the domestic horse and aims to consolidate current information on equine sleep, in relation to other species, in order to (a) identify both quantitatively and qualitatively what constitutes normal sleep in the horse, (b) identify optimal methods to measure equine sleep (logistically and in terms of accuracy), (c) determine whether changes in equine sleep quantity and quality reflect changes in the animal's welfare, and (d) recognize the primary factors that affect the quantity and quality of equine sleep. The review then discusses gaps in current knowledge and uses this information to identify and set the direction of future equine sleep research with the ultimate aim of improving equine performance and welfare. The conclusions from this review are also contextualized within the current discussions around the “social license” of horse use from a welfare perspective.
Collapse
Affiliation(s)
- Linda Greening
- Hartpury University and Hartpury College, Gloucester, United Kingdom
- *Correspondence: Linda Greening
| | - Sebastian McBride
- Institute of Biological, Environmental and Rural Science, Aberystwyth University, Aberystwyth, United Kingdom
| |
Collapse
|
13
|
Balikji J, Hoogbergen MM, Garssen J, Roth T, Verster JC. Insomnia Complaints and Perceived Immune Fitness in Young Adults with and without Self-Reported Impaired Wound Healing. Medicina (B Aires) 2022; 58:medicina58081049. [PMID: 36013516 PMCID: PMC9412748 DOI: 10.3390/medicina58081049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: Adequate sleep and an effective immune system are both essential to maintain a good health status. The current study aimed to determine the nature of insomnia complaints and perceived immune fitness among Dutch young adults with and without self-reported impaired wound healing. Materials and Methods: A total of (n = 2033) Dutch students (83.8% women) completed an online survey. Perceived immune fitness was assessed with a single-item scale and insomnia complaints with the SLEEP-50 insomnia subscale. The sample comprised a control group without self-reported impaired wound healing (n = 1622), a wound infection (WI) group (n = 69), a slow healing wounds (SHW) group (n = 250), and a COMBI group that experienced both WI and SHW (n = 92). Results: Comparisons with the control group revealed that individuals of the SHW and COMBI groups reported significantly poorer perceived immune functioning, increased insomnia complaints and daytime fatigue, and poorer sleep quality. Conclusions: Individuals with self-reported impaired wound healing have a poorer perceived immune functioning, increased insomnia complaints, daytime fatigue, and poorer sleep quality.
Collapse
Affiliation(s)
- Jessica Balikji
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Maarten M. Hoogbergen
- Division of Plastic Surgery, Catharina Ziekenhuis, Michelangelolaan 2, 5623 EJ Eindhoven, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Global Centre of Excellence Immunology, Nutricia Danone Research, 3584 CT Utrecht, The Netherlands
| | - Thomas Roth
- Sleep Disorders & Research Centre, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Joris C. Verster
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122, Australia
- Correspondence: ; Tel.: +31-30-253-6909
| |
Collapse
|
14
|
Neilson BD, Shepherd MH, Dickerson C, Chaconas EJ, Young JL, Rhon DI. Relationship Between Attitudes and Beliefs About Sleep, Sleep Disturbance, and Pain Interference in Patients With Spinal Pain. Clin J Pain 2022; 38:541-549. [PMID: 35642567 DOI: 10.1097/ajp.0000000000001051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Sleep impairments are a strong predictor of pain, making sleep a potential interest when treating patients with spine pain. Typical beliefs about the importance of sleep in patients seeking care for spinal pain are unknown. The purpose of this study was to describe the beliefs and attitudes about sleep in patients seeking care for spinal pain and to examine the relationships between dysfunctional beliefs and attitudes about sleep (DBAS), disordered sleep, and pain interference. MATERIALS AND METHODS This cross-sectional study included patients presenting to physical therapy with spine pain. Participants completed questionnaires including demographics, medical history, pain interference (pain, enjoyment, and general activity), DBAS-16, and sleep-related impairment (Patient-Reported Outcome Measurement Information System). Correlations were calculated between DBAS-16 scores and measures of sleep quality/quantity, and a generalized linear model was used to investigate the predictive ability of DBAS-16 scores on pain interference. RESULTS The mean DBAS-16 score was 4.22 (SD=2.03), with 52.5% of participants having DBAS. There was a strong relationship between DBAS-16 and Patient-Reported Outcome Measurement Information System ( rs =0.7; P <0.001). For every point higher score on the DBAS-16, pain interference scores increased by approximately half a point (B=0.46; 95% CI 0.33, 0.59, 1.80; P <0.001). DISCUSSION These results highlight a strong relationship between beliefs and attitudes about sleep and measures of sleep quality/quantity and a linear association with pain interference scores. These findings provide a rationale for targeting beliefs and attitudes about sleep when managing pain-related symptoms in patients seeking care for spine pain.
Collapse
Affiliation(s)
- Brett D Neilson
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
- Doctor of Physical Therapy Program, Hawai'i Pacific University, Honolulu, HI
| | - Mark H Shepherd
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
| | - Chris Dickerson
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
| | - Eric J Chaconas
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
| | - Jodi L Young
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
| | - Daniel I Rhon
- Doctor of Science in Physical Therapy Program, Bellin College, Green Bay, WI
- Department of Rehab Medicine, Uniformed Services University of Health Sciences, Bethesda, MD
| |
Collapse
|
15
|
Ragnoli B, Pochetti P, Pignatti P, Barbieri M, Mondini L, Ruggero L, Trotta L, Montuschi P, Malerba M. Sleep Deprivation, Immune Suppression and SARS-CoV-2 Infection. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:904. [PMID: 35055726 PMCID: PMC8775678 DOI: 10.3390/ijerph19020904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/18/2022]
Abstract
Sleep health and its adaptation to individual and environmental factors are crucial to promote physical and mental well-being across animal species. In recent years, increasing evidence has been reported regarding the relationship between sleep and the immune system and how sleep disturbances may perturb the delicate balance with severe repercussions on health outcomes. For instance, experimental sleep deprivation studies in vivo have reported several major detrimental effects on immune health, including induced failure of host defense in rats and increased risk for metabolic syndrome (MetS) and immune suppression in humans. In addition, two novel risk factors for dysregulated metabolic physiology have recently been identified: sleep disruption and circadian misalignment. In light of these recent findings about the interplay between sleep and the immune system, in this review, we focus on the relationship between sleep deprivation and immunity against viruses, with a special interest in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Beatrice Ragnoli
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Patrizia Pochetti
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS Pavia, 27100 Pavia, Italy;
| | - Mariangela Barbieri
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Lucrezia Mondini
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Luca Ruggero
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Liliana Trotta
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
| | - Paolo Montuschi
- Pharmacology Department, Faculty of Medicine, Catholic University of the Sacred Heart, 20123 Milan, Italy;
- Faculty of Medicine, National Hearth and Lung Institute, Imperial College of Science Technology and Medicine, Airways Disease Section, London SW7 2BX, UK
| | - Mario Malerba
- Respiratory Unit, S. Andrea Hospital, 13100 Vercelli, Italy; (B.R.); (P.P.); (M.B.); (L.M.); (L.R.); (L.T.)
- Department of Traslational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
16
|
Lam MTY, Malhotra A, LaBuzetta JN, Kamdar BB. Sleep in Critical Illness. Respir Med 2022. [DOI: 10.1007/978-3-030-93739-3_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Olekhnovich EI, Batotsyrenova EG, Yunes RA, Kashuro VA, Poluektova EU, Veselovsky VA, Ilina EN, Danilenko VN, Klimina KM. The effects of Levilactobacillus brevis on the physiological parameters and gut microbiota composition of rats subjected to desynchronosis. Microb Cell Fact 2021; 20:226. [PMID: 34930242 PMCID: PMC8686522 DOI: 10.1186/s12934-021-01716-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/02/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND All living organisms have developed during evolution complex time-keeping biological clocks that allowed them to stay attuned to their environments. Circadian rhythms cycle on a near 24 h clock. These encompass a variety of changes in the body ranging from blood hormone levels to metabolism, to the gut microbiota composition and others. The gut microbiota, in return, influences the host stress response and the physiological changes associated with it, which makes it an important determinant of health. Lactobacilli are traditionally consumed for their prophylactic and therapeutic benefits against various diseases, namely, the inflammatory bowel syndrome, and even emerged recently as promising psychobiotics. However, the potential role of lactobacilli in the normalization of circadian rhythms has not been addressed. RESULTS Two-month-old male rats were randomly divided into three groups and housed under three different light/dark cycles for three months: natural light, constant light and constant darkness. The strain Levilactobacillus brevis 47f was administered to rats at a dose of 0.5 ml per rat for one month and The rats were observed for the following two months. As a result, we identified the biomarkers associated with intake of L. brevis 47f. Changing the light regime for three months depleted the reserves of the main buffer in the cell-reduced glutathione. Intake of L. brevis 47f for 30 days restored cellular reserves of reduced glutathione and promoted redox balance. Our results indicate that the levels of urinary catecholamines correlated with light/dark cycles and were influenced by intake of L. brevis 47f. The gut microbiota of rats was also influenced by these factors. L. brevis 47f intake was associated with an increase in the relative abundance of Faecalibacterium and Roseburia and a decrease in the relative abundance of Prevotella and Bacteroides. CONCLUSIONS The results of this study show that oral administration of L. brevis 47f, for one month, to rats housed under abnormal lightning conditions (constant light or constant darkness) normalized their physiological parameters and promoted the gut microbiome's balance.
Collapse
Affiliation(s)
- Evgenii I. Olekhnovich
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435 Russia
| | - Ekaterina G. Batotsyrenova
- Saint Petersburg State Pediatric Medical University, 2 Litovskaya str., St. Petersburg, 194100 Russia
- Golikov Research Center of Toxicology Under Federal Medical Biological Agency, 1 Bekhtereva str., St. Petersburg, 192019 Russia
| | - Roman A. Yunes
- Department of Genetics and Biotechnology, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, 119991 Russia
| | - Vadim A. Kashuro
- Saint Petersburg State Pediatric Medical University, 2 Litovskaya str., St. Petersburg, 194100 Russia
- Golikov Research Center of Toxicology Under Federal Medical Biological Agency, 1 Bekhtereva str., St. Petersburg, 192019 Russia
| | - Elena U. Poluektova
- Department of Genetics and Biotechnology, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, 119991 Russia
| | - Vladimir A. Veselovsky
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435 Russia
| | - Elena N. Ilina
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435 Russia
| | - Valeriy N. Danilenko
- Department of Genetics and Biotechnology, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, 119991 Russia
| | - Ksenia M. Klimina
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435 Russia
- Department of Genetics and Biotechnology, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, 119991 Russia
| |
Collapse
|
18
|
Differential Effect of Light and Dark Period Sleep Fragmentation on Composition of Gut Microbiome and Inflammation in Mice. Life (Basel) 2021; 11:life11121283. [PMID: 34947814 PMCID: PMC8709399 DOI: 10.3390/life11121283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022] Open
Abstract
Bi-directional interactions amongst the gut microbiota, immune system, and brain function are thought to be critical mediators of health and disease. The role sleep plays in mediating these interactions is not known. We assessed the effects of sleep fragmentation (SF) on the microbiota–gut–brain axis. Male C57BL/6NCrl mice (4 to 5 per cage, fed standard lab chow) experienced SF via mechanical stimulation at 2 min intervals during the light (SF) and dark (DD, dark disturbances) periods. Home cage (HC) controls were undisturbed. After 10 days, fecal samples were collected at light onset, midday, light offset, and midnight. Samples were also collected after 10 days without SF. Subsequently, the mice were randomized across groups and allowed 20 additional days of recovery followed by 10 days of SF or DD. To assess effects on the microbiota, 16S rRNA sequencing was used, and mesenteric lymph nodes (MLNs) and cortex and medial prefrontal cortex were analyzed using cytokine arrays. SF and DD produced significant alterations in the microbiota compared to HC, and DD had greater impact than SF on some organisms. SF produced marked suppression in MLNs of chemokines that regulate inflammation (CCL3, CCL4 and their receptor CCR5) and maintain the immune mucosal barrier (Cxcl13) at the same time that cortical cytokines (IL-33) indicated neuroinflammation. DD effects on immune responses were similar to HC. These data suggest that SF alters the microbiome and suppresses mucosal immunity at the same time that mediators of brain inflammation are upregulated. The translational implications for potential application to clinical care are compelling.
Collapse
|
19
|
Bonilla-Jaime H, Zeleke H, Rojas A, Espinosa-Garcia C. Sleep Disruption Worsens Seizures: Neuroinflammation as a Potential Mechanistic Link. Int J Mol Sci 2021; 22:12531. [PMID: 34830412 PMCID: PMC8617844 DOI: 10.3390/ijms222212531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances, such as insomnia, obstructive sleep apnea, and daytime sleepiness, are common in people diagnosed with epilepsy. These disturbances can be attributed to nocturnal seizures, psychosocial factors, and/or the use of anti-epileptic drugs with sleep-modifying side effects. Epilepsy patients with poor sleep quality have intensified seizure frequency and disease progression compared to their well-rested counterparts. A better understanding of the complex relationship between sleep and epilepsy is needed, since approximately 20% of seizures and more than 90% of sudden unexpected deaths in epilepsy occur during sleep. Emerging studies suggest that neuroinflammation, (e.g., the CNS immune response characterized by the change in expression of inflammatory mediators and glial activation) may be a potential link between sleep deprivation and seizures. Here, we review the mechanisms by which sleep deprivation induces neuroinflammation and propose that neuroinflammation synergizes with seizure activity to worsen neurodegeneration in the epileptic brain. Additionally, we highlight the relevance of sleep interventions, often overlooked by physicians, to manage seizures, prevent epilepsy-related mortality, and improve quality of life.
Collapse
Affiliation(s)
- Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Área de Biología Conductual y Reproductiva, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico CP 09340, Mexico;
| | - Helena Zeleke
- Neuroscience and Behavioral Biology Program, College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA;
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Claudia Espinosa-Garcia
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
20
|
Garbarino S, Lanteri P, Bragazzi NL, Magnavita N, Scoditti E. Role of sleep deprivation in immune-related disease risk and outcomes. Commun Biol 2021; 4:1304. [PMID: 34795404 PMCID: PMC8602722 DOI: 10.1038/s42003-021-02825-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Modern societies are experiencing an increasing trend of reduced sleep duration, with nocturnal sleeping time below the recommended ranges for health. Epidemiological and laboratory studies have demonstrated detrimental effects of sleep deprivation on health. Sleep exerts an immune-supportive function, promoting host defense against infection and inflammatory insults. Sleep deprivation has been associated with alterations of innate and adaptive immune parameters, leading to a chronic inflammatory state and an increased risk for infectious/inflammatory pathologies, including cardiometabolic, neoplastic, autoimmune and neurodegenerative diseases. Here, we review recent advancements on the immune responses to sleep deprivation as evidenced by experimental and epidemiological studies, the pathophysiology, and the role for the sleep deprivation-induced immune changes in increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still remain. Further understanding of the causal relationship between sleep deprivation and immune deregulation would help to identify individuals at risk for disease and to prevent adverse health outcomes.
Collapse
Affiliation(s)
- Sergio Garbarino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, 16132, Genoa, Italy.
| | - Paola Lanteri
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, M3J 1P3, Canada
| | - Nicola Magnavita
- Postgraduate School of Occupational Medicine, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Department of Woman/Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100, Lecce, Italy
| |
Collapse
|
21
|
Abstract
A molecular circadian clock exists not only in the brain, but also in most cells of the body. Research over the past two decades has demonstrated that it directs daily rhythmicity of nearly every aspect of metabolism. It also consolidates sleep-wake behavior each day into an activity/feeding period and a sleep/fasting period. Otherwise, sleep-wake states are mostly controlled by hypothalamic and thalamic regulatory circuits in the brain that direct overall brain state. Recent evidence suggests that hypothalamic control of appetite and metabolism may be concomitant with sleep-wake regulation, and even share the same control centers. Thus, circadian control of metabolic pathways might be overlaid by sleep-wake control of the same pathways, providing a flexible and redundant system to modify metabolism according to both activity and environment.
Collapse
|
22
|
O’Brien KE, Riddell NE, Gómez-Olivé FX, Rae DE, Scheuermaier K, von Schantz M. Sleep Disturbances in HIV Infection and their Biological Basis. Sleep Med Rev 2021; 65:101571. [DOI: 10.1016/j.smrv.2021.101571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
|
23
|
Panchin Y, Kovalzon VM. Total Wake: Natural, Pathological, and Experimental Limits to Sleep Reduction. Front Neurosci 2021; 15:643496. [PMID: 33897357 PMCID: PMC8058214 DOI: 10.3389/fnins.2021.643496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/26/2021] [Indexed: 11/16/2022] Open
Abstract
Sleep is not considered a pathological state, but it consumes a third of conscious human life. This share is much more than most optimistic life extension forecasts that biotechnologies or experimental and medical interventions can offer. Are there insurmountable physical or biological limitations to reducing the duration of sleep? How far can it be avoided without fatal consequences? What means can reduce the length of sleep? It is widely accepted that sleep is necessary for long-term survival. Here we review the limited yet intriguing evidence that is not consistent with this notion. We concentrate on clinical cases of complete and partial loss of sleep and on human mutations that result in a short sleep phenotype. These observations are supported by new animal studies and are discussed from the perspective of sleep evolution. Two separate hypotheses suggest distinct approaches for remodeling our sleep machinery. If sleep serves an unidentified vital physiological function, this indispensable function has to be identified before "sleep prosthesis" (technical, biological, or chemical) can be developed. If sleep has no vital function, but rather represents a timing mechanism for adaptive inactivity, sleep could be reduced by forging the sleep generation system itself, with no adverse effects.
Collapse
Affiliation(s)
- Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Department of Mathematical Methods in Biology, Belozersky Institute, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir M. Kovalzon
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
24
|
Ruiz FS, Rosa DS, Zimberg IZ, Dos Santos Quaresma MV, Nunes JO, Apostolico JS, Weckx LY, Souza AR, Narciso FV, Fernandes-Junior SA, Gonçalves B, Folkard S, Bittencourt L, Tufik S, Tulio de Mello M. Night shift work and immune response to the meningococcal conjugate vaccine in healthy workers: a proof of concept study. Sleep Med 2020; 75:263-275. [PMID: 32866895 DOI: 10.1016/j.sleep.2020.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND It is well-established that sleep regulates immune functions. Immunological functions are dependent on circadian rhythms and regular sleep as both have an impact on the magnitude of immune responses following antigenic challenge (eg, in vaccination). Here we investigated whether nocturnal shift work can influence post-vaccination response. METHODS Thirty-four healthy workers (23 females) working either nocturnal or diurnal shifts (17 in each group) received the meningococcal C meningitis vaccine. Sleep was recorded polysomnographically (PSG) and with actigraphy. Humoral and cellular responses were assessed after vaccination. RESULTS Night workers showed decreased N3 stage and REM sleep duration, increased inflammatory mediators (TNF-α and IL-6 levels), and a weak specific humoral response to vaccination associated with reduced CD4 T lymphocytes, reduced plasmacytoid dendritic cells, reduced prolactin levels, increased TReg and increased IL-10 levels. In addition, the decrease in total sleep time and circadian rhythm alterations were associated with a reduced humoral response post-vaccination. CONCLUSIONS Our findings provide novel evidence concerning immune alterations of shift work on workers' health based on real-life circumstances. In association with circadian components, sufficient sleep time and rhythm synchronization were important for the development of the Ag-specific immune response, suggesting that the humoral response to vaccination may be impaired in individuals with chronic sleep restriction and circadian misalignment.
Collapse
Affiliation(s)
- Francieli S Ruiz
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK.
| | - Daniela S Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Ioná Z Zimberg
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Jethe Of Nunes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Juliana S Apostolico
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Lily Y Weckx
- Departamento de Pediatria, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Alessandra R Souza
- Departamento de Pediatria, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fernanda V Narciso
- Escola de Educação Física, Fisioterapia e Terapia Ocupacional (EEFFTO), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Bruno Gonçalves
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Brazil
| | | | - Lia Bittencourt
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marco Tulio de Mello
- Escola de Educação Física, Fisioterapia e Terapia Ocupacional (EEFFTO), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
25
|
Suganya K, Kayalvizhi E, Yuvaraj R, Chandrasekar M, Kavitha U, Konakanchi Suresh K. Effect of Withania Somnifera on the antioxidant and neurotransmitter status in sleep deprivation induced Wistar rats. Bioinformation 2020; 16:631-637. [PMID: 33214752 PMCID: PMC7649022 DOI: 10.6026/97320630016631] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/31/2020] [Indexed: 11/23/2022] Open
Abstract
Sleep is normally a period of relaxation and repair, important for the maintenance of physiological homeostasis and psychological balance. "Globally, millions of people experiences sleep deprivation daily". Sleep deprivation (SD) impairs cognitive functions, decreases anti-oxidative defense and induces neuronal changes. Withania somnifera (WS), commonly known as an "Indian Ginseng" has broad therapeutic applications, including anti-inflammatory activities, actions on immune system, circulatory system, central nervous system etc., The study is aimed to assess effect of Withania somnifera on antioxidant status and neurotransmitter level in sleep deprivation induced male Wistar albino rats. The study was done in the Department of Physiology, Meenakshi Medical College and Hospital, Enathur, Kanchipuram. 24 male adult Wistar rats weighing 120-150g were used for the study. They were divided into 4 groups with 6 animals in each group. (Group I - cage control, Group II - large platform control, Group III - sleep deprived group and Group IV - WS treated SD group). Animals were deprived sleep for one week using a modified multiple platform method. Oxidative stress parameters and antioxidant enzymes were measured using spectrophotometry. Neurotransmitters such as dopamine and serotonin concentration in the serum were measured by ELISA method. There was a marked (by one-way ANOVA test) decrease observed in the antioxidants enzymes in the cortex of both large platform control and sleep deprivation induced group. The group treated with W. somnifera root extract significantly reduced the free radical production and lipid peroxidation with simultaneous increase in the level of antioxidant enzymes compared to the untreated group. Also in our study the concentration of dopamine and serotonin was found to be significantly reduced (p < 0.05) in sleep deprived (SD) and large platform control group when compared to cage control group. Whereas the group treated with W. somnifera (400mg/kg b.wt) increased the neurotransmitter levels significantly. Withania somnifera proved to be an effective therapeutic agent by maintaining the antioxidant status and neurotransmitter levels.
Collapse
Affiliation(s)
- K Suganya
- Meenakshi Medical College Hospital and RI, Physiology, India
| | - E Kayalvizhi
- Meenakshi Medical College Hospital and RI, Physiology, India
| | | | | | - U Kavitha
- Meenakshi Medical College Hospital and RI, Physiology, India
| | | |
Collapse
|
26
|
Mehta R, Giri S, Mallick BN. REM sleep loss-induced elevated noradrenaline could predispose an individual to psychosomatic disorders: a review focused on proposal for prediction, prevention, and personalized treatment. EPMA J 2020; 11:529-549. [PMID: 33240449 DOI: 10.1007/s13167-020-00222-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
Historically and traditionally, it is known that sleep helps in maintaining healthy living. Its duration varies not only among individuals but also in the same individual depending on circumstances, suggesting it is a dynamic and personalized physiological process. It has been divided into rapid eye movement sleep (REMS) and non-REMS (NREMS). The former is unique that adult humans spend the least time in this stage, when although one is physically asleep, the brain behaves as if awake, the dream state. As NREMS is a pre-requisite for appearance of REMS, the latter can be considered a predictive readout of sleep quality and health. It plays a protective role against oxidative, stressful, and psychopathological insults. Several modern lifestyle activities compromise quality and quantity of sleep (including REMS) affecting fundamental physiological and psychopathosomatic processes in a personalized manner. REMS loss-induced elevated brain noradrenaline (NA) causes many associated symptoms, which are ameliorated by preventing NA action. Therefore, we propose that awareness about personalized sleep hygiene (including REMS) and maintaining optimum brain NA level should be of paramount significance for leading physical and mental well-being as well as healthy living. As sleep is a dynamic, multifactorial, homeostatically regulated process, for healthy living, we recommend addressing and treating sleep dysfunctions in a personalized manner by the health professionals, caregivers, family, and other supporting members in the society. We also recommend that maintaining sleep profile, optimum level of NA, and/or prevention of elevation of NA or its action in the brain must be seriously considered for ameliorating lifestyle and REMS disturbance-associated dysfunctions.
Collapse
Affiliation(s)
- Rachna Mehta
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India.,Present Address: Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Shatrunjai Giri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India
| | - Birendra N Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India
| |
Collapse
|
27
|
Jiang X, Zheng J, Zhang S, Wang B, Wu C, Guo X. Advances in the Involvement of Gut Microbiota in Pathophysiology of NAFLD. Front Med (Lausanne) 2020; 7:361. [PMID: 32850884 PMCID: PMC7403443 DOI: 10.3389/fmed.2020.00361] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis and progresses to non-steatohepatitis (NASH) when the liver displays overt inflammatory damage. Increasing evidence has implicated critical roles for dysbiosis and microbiota-host interactions in NAFLD pathophysiology. In particular, microbiota alter intestine absorption of nutrients and intestine permeability, whose dysregulation enhances the delivery of nutrients, endotoxin, and microbiota metabolites to the liver and exacerbates hepatic fat deposition and inflammation. While how altered composition of gut microbiota attributes to NAFLD remains to be elucidated, microbiota metabolites are shown to be involved in the regulation of hepatocyte fat metabolism and liver inflammatory responses. In addition, intestinal microbes and circadian coordinately adjust metabolic regulation in different stages of life. During aging, altered composition of gut microbiota, along with circadian clock dysregulation, appears to contribute to increased incidence and/or severity of NAFLD.
Collapse
Affiliation(s)
- Xiaofan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shixiu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baozhen Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
28
|
Triplett J, Ellis D, Braddock A, Roberts E, Ingram K, Perez E, Short A, Brown D, Hutzley V, Webb C, Soto A, Chan V. Temporal and region-specific effects of sleep fragmentation on gut microbiota and intestinal morphology in Sprague Dawley rats. Gut Microbes 2020; 11:706-720. [PMID: 31924109 PMCID: PMC7524289 DOI: 10.1080/19490976.2019.1701352] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Sleep is a fundamental biological process, that when repeatedly disrupted, can result in severe health consequences. Recent studies suggest that both sleep fragmentation (SF) and dysbiosis of the gut microbiome can lead to metabolic disorders, though the underlying mechanisms are largely unclear. To better understand the consequences of SF, we investigated the effects of acute (6 days) and chronic (6 weeks) SF on rats by examining taxonomic profiles of microbiota in the distal ileum, cecum and proximal colon, as well as assessing structural and functional integrity of the gastrointestinal barrier. We further assayed the impact of SF on a host function by evaluating inflammation and immune response. Both acute and chronic SF induced microbial dysbiosis, more dramatically in the distal ileum (compared to other two regions studied), as noted by significant perturbations in alpha- and beta-diversity; though, specific microbial populations were significantly altered throughout each of the three regions. Furthermore, chronic SF resulted in increased crypt depth in the distal ileum and an increase in the number of villi lining both the cecum and proximal colon. Additional changes were noted with chronic SF, including: decreased microbial adhesion and penetration in the distal ileum and cecum, elevation in serum levels of the cytokine KC/GRO, and depressed levels of corticotropin. Importantly, our data show that perturbations to microbial ecology and intestinal morphology intensify in response to prolonged SF and these changes are habitat specific. Together, these results reveal consequences to gut microbiota homeostasis and host response following acute and chronic SF in rats.
Collapse
Affiliation(s)
- Judy Triplett
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - David Ellis
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amber Braddock
- Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Wright-Patterson AFB, OH, USA
| | - Erin Roberts
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Katherine Ingram
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Eric Perez
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amanda Short
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Dominique Brown
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victoria Hutzley
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Chelsey Webb
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Armando Soto
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victor Chan
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA,CONTACT Victor Chan Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| |
Collapse
|
29
|
Hahn J, Günter M, Schuhmacher J, Bieber K, Pöschel S, Schütz M, Engelhardt B, Oster H, Sina C, Lange T, Autenrieth SE. Sleep enhances numbers and function of monocytes and improves bacterial infection outcome in mice. Brain Behav Immun 2020; 87:329-338. [PMID: 31904407 DOI: 10.1016/j.bbi.2020.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/15/2019] [Accepted: 01/01/2020] [Indexed: 01/21/2023] Open
Abstract
Sleep strongly impacts both humoral and cellular immunity; however, its acute effects on the innate immune defense against pathogens are unclear. Here, we elucidated in mice whether sleep affects the numbers and functions of innate immune cells and their defense against systemic bacterial infection. Sleep significantly increased numbers of classical monocytes in blood and spleen of mice that were allowed to sleep for six hours at the beginning of the normal resting phase compared to mice kept awake for the same time. The sleep-induced effect on classical monocytes was neither caused by alterations in corticosterone nor myelopoiesis, bone marrow egress or death of monocytes and did only partially involve Gαi-protein coupled receptors like chemokine receptor 2 (CCR2), but not the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) or lymphocyte function-associated antigen 1 (LFA-1). Notably, sleep suppressed the expression of the clock gene Arntl in splenic monocytes and the sleep-induced increase in circulating classical monocytes was abrogated in Arntl-deficient animals, indicating that sleep is a prerequisite for clock-gene driven rhythmic trafficking of classical monocytes. Sleep also enhanced the production of reactive oxygen species by monocytes and neutrophils. Moreover, sleep profoundly reduced bacterial load in blood and spleen of mice that were allowed to sleep before systemic bacterial infection and consequently increased survival upon infection. These data provide the first evidence that sleep enhances numbers and function of innate immune cells and therewith strengthens early defense against bacterial pathogens.
Collapse
Affiliation(s)
- Julia Hahn
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Manina Günter
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Juliane Schuhmacher
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Kristin Bieber
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - Simone Pöschel
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - Monika Schütz
- Institute for Medical Microbiology and Hygiene, University of Tübingen, 72076 Tübingen, Germany
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany
| | - Christian Sina
- Institute for Nutritional Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Tanja Lange
- Department of Rheumatology & Clinical Immunology, University of Lübeck, 23562 Lübeck, Germany
| | - Stella E Autenrieth
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
30
|
Ayadilord M, Mahmoudzadeh S, Hoseini ZS, Askari M, Rezapour H, Saharkhiz M, Abbaszadeh A, Karbasi S, Zandi Dashtebayaze N, Ferns GA, Bahrami A. Neuropsychological function is related to irritable bowel syndrome in women with premenstrual syndrome and dysmenorrhea. Arch Gynecol Obstet 2020; 302:915-923. [PMID: 32594296 DOI: 10.1007/s00404-020-05659-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is increasing evidence demonstrating the co-occurrence of primary dysmenorrhea (PD), premenstrual syndrome (PMS), and irritable bowel syndrome (IBS) in women. This study aimed to investigate whether women who have symptoms of IBS in addition to PD and PMS also report more severe or frequent menstruation-associated symptoms and psychological complications compared to women with PD and PMS alone. METHODS The study group included 182 female University students aged 18-25 years. IBS was diagnosed using the Rome III criteria. The severity of PMS and PD was determined using a 10-point visual analog scale and PSST (Premenstrual Syndrome Screening Tool), respectively. Neuropsychological functions including cognitive function, depression score, anxiety score, stress, insomnia, daytime sleepiness, quality of life and personality were assessed using standard questionnaires. RESULTS Of the 182 young females, 31 (17.0%) had IBS. Average days of bleeding during the menstrual cycle and mean pain severity on the PSST scale were significantly greater in the group with IBS compared to the non-IBS group (p < 0.01). The non-IBS individuals scored more favorably than the women with IBS with respect to severity of depression, insomnia, daytime sleepiness (p < 0.05). The PSST scores were significantly correlated with scores for depression (r = 0.29; p < 0.001), anxiety (r = 0.28; p < 0.001), stress (r = 0.32; p < 0.001), insomnia (r = 0.34; p < 0.001) and daytime sleepiness (r = 0.31; p < 0.001); while, they were negatively correlated with cognitive abilities (r = - 0.20; p = 0.006) and quality of life (r = - 0.42; p < 0.001). Linear regression analysis showed that the PSST scores were possibly significant factors in determining the scores for depression, anxiety, stress, quality of life, insomnia and daytime sleepiness (p < 0.05). CONCLUSION IBS is related to psychological comorbidities, in particular depression, sleep problems and menstrual-associated disorders. IBS may exacerbate the features of PMS which should be taken into account in the management of PMS.
Collapse
Affiliation(s)
- Malaksima Ayadilord
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Sara Mahmoudzadeh
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Masoumeh Askari
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Hadis Rezapour
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mansoore Saharkhiz
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Arefeh Abbaszadeh
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Samira Karbasi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
31
|
Matenchuk BA, Mandhane PJ, Kozyrskyj AL. Sleep, circadian rhythm, and gut microbiota. Sleep Med Rev 2020; 53:101340. [PMID: 32668369 DOI: 10.1016/j.smrv.2020.101340] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022]
Abstract
From asthma and heart disease to diabetes and obesity, the human microbiome plays a role in the pathogenesis of each chronic health condition plaguing today's society. Recent work has shown that the gut microbiota and its metabolites exhibit diurnal rhythmicity which predominantly respond to the feeding/fasting cycle. Persistent jet lag, an obesogenic diet, and clock gene deficiency can dampen the oscillatory nature of gut bacterial composition, which can subsequently be rescued by time restricted feeding. Contrastingly, gut microbial metabolites influence central and hepatic clock gene expression and sleep duration in the host and regulate body composition through circadian transcription factors. Both sleep fragmentation and short sleep duration are associated with gut dysbiosis which may be due to activation of the HPA-axis. Metabolic disturbances associated with sleep loss may in fact be mediated through the overgrowth of specific gut bacteria. Reciprocally, the end products of bacterial species which grow in response to sleep loss are able to induce fatigue. Furthermore, probiotic supplementation has been found to improve subjective sleep quality. Sleep quality and duration may be an important target for supporting healthy gut microbiota composition, but the cyclic nature of this relationship should not be overlooked.
Collapse
Affiliation(s)
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
32
|
Oles V, Koh KMS, Dykstra-Aiello CJ, Savenkova M, Gibbons CM, Nguyen JT, Karatsoreos I, Panchenko A, Krueger JM. Sleep- and time of day-linked RNA transcript expression in wild-type and IL1 receptor accessory protein-null mice. J Appl Physiol (1985) 2020; 128:1506-1522. [PMID: 32324480 DOI: 10.1152/japplphysiol.00839.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sleep regulation involves interleukin-1β (IL1) family members, TNF, and circadian clock genes. Previously, we characterized spontaneous sleep and sleep after 8 h of sleep deprivation (SD) ending at zeitgeber time (ZT)4 and ZT16 in wild-type (WT) and IL1 receptor accessory protein (AcP)- and brain-specific AcP (AcPb)-knockout (KO) mice. Here, we applied quantitative reverse transcriptase polymerase chain reaction and Spearman gene pair expression correlation methods to characterize IL1, IL1 receptor 1 (IL1R1), AcP, AcPb, Period 1 (Per1), Clock, adenosine deaminase (Ada), peptidoglycan recognition protein 1 (Pglyrp1), and TNF mRNA expressions under conditions with distinct sleep phenotypes. In WT mice, IL1, IL1R1, AcP, Ada, and Clock mRNAs were higher at ZT4 (mid-sleep period) than at ZT16. mRNA expressions differed substantially in AcP and AcPb KO mice at those times. After SD ending at ZT4, only WT mice had a non-rapid eye movement sleep (NREMS) rebound, and AcPb and IL1R1 mRNA increases were unique to WT mice. In AcPb KO mice, which have spontaneous high EEG slow wave power, AcP and Pglyrp1 mRNAs were elevated relative to WT mice at ZT4. At ZT4, the AcPb KO - WT Spearman correlation difference networks showed high positive correlations between IL1R1 and IL1, Per1, and Clock and high negative correlations between TNF and Pglyrp1 and Ada. At ZT16, the WT mice gene pair expression network was mostly negative, whereas in AcP KO mice, which have substantially more rapid eye movement sleep than WT mice, it was all positive. We conclude that gene pair expression correlations depend on the presence of AcP and AcPb.NEW & NOTEWORTHY Spearman gene pair expression correlations depend upon the presence or absence of interleukin-1 receptor accessory protein and upon sleep phenotype.
Collapse
Affiliation(s)
- Vladyslav Oles
- Department of Mathematics and Statistics, Washington State University, Pullman, Washington
| | - Khia Min Sabrina Koh
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | | | - Marina Savenkova
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Cody M Gibbons
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington.,University of Washington School of Medicine, Seattle, Washington
| | - Joseph T Nguyen
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Ilia Karatsoreos
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Alexander Panchenko
- Department of Mathematics and Statistics, Washington State University, Pullman, Washington
| | - James M Krueger
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
33
|
Wang G, Wu X, Zhu G, Han S, Zhang J. Dexmedetomidine alleviates sleep-restriction-mediated exaggeration of postoperative immunosuppression via splenic TFF2 in aged mice. Aging (Albany NY) 2020; 12:5318-5335. [PMID: 32200357 PMCID: PMC7138555 DOI: 10.18632/aging.102952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Guangzhi Wang
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou 450003, Henan, China
| | - Xiaoying Wu
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou 450003, Henan, China
| | - Guosong Zhu
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou 450003, Henan, China
| | - Shuangyin Han
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou 450003, Henan, China
| | - Jiaqiang Zhang
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou 450003, Henan, China
| |
Collapse
|
34
|
Garofalo S, Picard K, Limatola C, Nadjar A, Pascual O, Tremblay MÈ. Role of Glia in the Regulation of Sleep in Health and Disease. Compr Physiol 2020; 10:687-712. [PMID: 32163207 DOI: 10.1002/cphy.c190022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep is a naturally occurring physiological state that is required to sustain physical and mental health. Traditionally viewed as strictly regulated by top-down control mechanisms, sleep is now known to also originate locally. Glial cells are emerging as important contributors to the regulation of sleep-wake cycles, locally and among dedicated neural circuits. A few pioneering studies revealed that astrocytes and microglia may influence sleep pressure, duration as well as intensity, but the precise involvement of these two glial cells in the regulation of sleep remains to be fully addressed, across contexts of health and disease. In this overview article, we will first summarize the literature pertaining to the role of astrocytes and microglia in the regulation of sleep under normal physiological conditions. Afterward, we will discuss the beneficial and deleterious consequences of glia-mediated neuroinflammation, whether it is acute, or chronic and associated with brain diseases, on the regulation of sleep. Sleep disturbances are a main comorbidity in neurodegenerative diseases, and in several brain diseases that include pain, epilepsy, and cancer. Identifying the relationships between glia-mediated neuroinflammation, sleep-wake rhythm disruption and brain diseases may have important implications for the treatment of several disorders. © 2020 American Physiological Society. Compr Physiol 10:687-712, 2020.
Collapse
Affiliation(s)
- Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Katherine Picard
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Agnès Nadjar
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France
| | - Olivier Pascual
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Université Claude Bernard Lyon, Lyon, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada.,Départment de médecine moleculaire, Faculté de médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
35
|
Kuvat N, Tanriverdi H, Armutcu F. The relationship between obstructive sleep apnea syndrome and obesity: A new perspective on the pathogenesis in terms of organ crosstalk. CLINICAL RESPIRATORY JOURNAL 2020; 14:595-604. [PMID: 32112481 DOI: 10.1111/crj.13175] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/13/2020] [Accepted: 02/23/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Obstructive sleep apnea syndrome (OSAS) is a common disorder that has a major impact on public health. The connection between OSAS and obesity is very complex and likely represents an interaction between biological and lifestyle factors. Oxidative stress, inflammation and metabolic dysregulation are both actors involved in the pathogenesis of OSAS and obesity. Also, the current evidence suggests that gut microbiota plays a significant role in the emergence and progression of some metabolic disorders. When the relationship between OSAS and obesity is evaluated extensively, it is understood that they show mutual causality with each other, and that metabolic challenges such as impaired microbiota affect this bidirectional organ interaction, and by ensuing organ injury. OBJECTIVES The aim of this study is to investigate the association between OSAS and obesity, and the effect of "organ crosstalk" on the pathogenesis of the relationship and to contribute to the diagnosis and treatment options in the light of current data. DATA SOURCE We performed an electronic database search including PubMed, EMBASE and Web of Science. We used the following search terms: OSAS, obesity, inflammation, metabolic dysregulation and gut microbiota. CONCLUSION Obesity and OSAS adversely affect many organs and systems. Besides the factors affecting the diagnosis of the OSAS-obesity relationship, mutual organ interactions among the respiratory system, adipose tissue and intestines should not be ignored for prevention and treatment of OSAS and obesity. Comprehensive clinical trials addressing the efficacy and efficiency of current or potential treatments on therapeutic applications in the OSAS-obesity relationship are needed.
Collapse
Affiliation(s)
- Nuray Kuvat
- Infectious Diseases and Clinical Microbiology, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Hakan Tanriverdi
- Department of Chest Diseases, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Ferah Armutcu
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
36
|
Szentirmai É, Kapás L. Sleep and body temperature in TNFα knockout mice: The effects of sleep deprivation, β3-AR stimulation and exogenous TNFα. Brain Behav Immun 2019; 81:260-271. [PMID: 31220563 PMCID: PMC6754767 DOI: 10.1016/j.bbi.2019.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 01/09/2023] Open
Abstract
Increased production of pro-inflammatory cytokines is assumed to mediate increased sleep under inflammatory conditions, such as systemic infections or recovery from sleep loss. The role of cytokines in sleep regulation under normal conditions is less clear. In the present study, we investigated the role of endogenous tumor necrosis factor alpha (TNFα) in sleep regulation using TNFα knockout (KO) mice. Under control conditions at thermoneutral ambient temperature, total sleep time did not differ between TNFα KO and wild-type (WT) mice, but TNFα KO mice had increased rapid-eye-movement sleep (REMS), accompanied by decreased motor activity and body temperature. Exposure to 17 °C induced decreases in total sleep time similarly in both genotypes. Sleep deprivation by gentle handling elicited robust rebound increases in non-rapid-eye movement sleep (NREMS), REMS and electroencephalographic (EEG) slow-wave activity (SWA), accompanied by suppressed motor activity and decreased body temperature; there was no significant difference between the responses of WT and KO mice. Systemic injection of the beta3-adrenergic receptor (β3-AR) agonist CL-316,243 induced increases in NREMS and body temperature. The temperature response, but not the sleep effect, was attenuated in the KO animals. Systemic injection of TNFα induced increased NREMS, reduced REMS and biphasic temperature responses in both genotypes. In the KO mice, the NREMS-promoting effects of exogenously administered TNFα was decreased, while REMS suppression was enhanced, and the first, hypothermic, phase of temperature response was attenuated. Overall, TNFα KO mice did not show any deficiency in sleep regulation which suggests that the role of endogenous TNFα in sleep regulation is less pronounced than previously suggested.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
37
|
Kiecolt-Glaser JK, Wilson SJ, Madison A. Marriage and Gut (Microbiome) Feelings: Tracing Novel Dyadic Pathways to Accelerated Aging. Psychosom Med 2019; 81:704-710. [PMID: 30308579 PMCID: PMC6458105 DOI: 10.1097/psy.0000000000000647] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Within a couple, partners influence each other's mental and physical health. This review focuses on how couples' relationships, the partners' individual and joint vulnerabilities, and their health behaviors influence health through changes in the gut microbiota, metabolism, and immune function. Couples' shared stressors and emotions and their intertwined lifestyles and routines serve to promote common disease risks in part through parallel changes in their gut microbiotas. Marital discord, stress, and depression have strong bidirectional links, fueling one another. Chronic marital stress and depression can elevate the risk for obesity, metabolic syndrome, and cardiovascular disease by altering resting energy expenditure, insulin production, and triglyceride responses after unhealthy meals. During stressful times, health behaviors typically suffer-and sleep disturbances, poor diets, and sedentary behavior all influence these metabolic pathways while also promoting gut dysbiosis. Dysbiosis increases intestinal permeability (gut leakiness), providing a mechanistic pathway from marital distress and depression to heightened inflammation and accelerated aging. Age-related changes in the gut microbiota's composition and gut leakiness foster immunosenescence, as well as the progression of inflamm-aging; these age-related risks may be altered by stress and depression, diet, sleep, exercise habits, and developmental shifts in emotion regulation strategies. Consideration of the strong mutual influences that partners have on each other's mood and health behaviors, as well as the biological pathways that underlie these influences, provides a new way to view marriage's health implications.
Collapse
Affiliation(s)
- Janice K. Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine
- Department of Psychiatry & Behavioral Health, The Ohio State University College of Medicine
| | - Stephanie J. Wilson
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine
| | - Annelise Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine
- Department of Psychology, The Ohio State University
| |
Collapse
|
38
|
The Role of Gut Microbiota in Intestinal Inflammation with Respect to Diet and Extrinsic Stressors. Microorganisms 2019; 7:microorganisms7080271. [PMID: 31430948 PMCID: PMC6722800 DOI: 10.3390/microorganisms7080271] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota maintains a symbiotic relationship with the host and regulates several important functions including host metabolism, immunity, and intestinal barrier function. Intestinal inflammation and inflammatory bowel disease (IBD) are commonly associated with dysbiosis of the gut microbiota. Alterations in the gut microbiota and associated changes in metabolites as well as disruptions in the intestinal barrier are evidence of the relationship between the gut microbiota and intestinal inflammation. Recent studies have found that many factors may alter the gut microbiota, with the effects of diet being commonly-studied. Extrinsic stressors, including environmental stressors, antibiotic exposure, sleep disturbance, physical activity, and psychological stress, may also play important roles in altering the composition of the gut microbiota. Herein, we discuss the roles of the gut microbiota in intestinal inflammation in relation to diet and other extrinsic stressors.
Collapse
|
39
|
Besedovsky L, Lange T, Haack M. The Sleep-Immune Crosstalk in Health and Disease. Physiol Rev 2019; 99:1325-1380. [PMID: 30920354 PMCID: PMC6689741 DOI: 10.1152/physrev.00010.2018] [Citation(s) in RCA: 671] [Impact Index Per Article: 134.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/08/2023] Open
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.
Collapse
Affiliation(s)
- Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Tanja Lange
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Monika Haack
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
40
|
Lasselin J, Ingre M, Regenbogen C, Olsson MJ, Garke M, Brytting M, Edgar R, Lekander M, Axelsson J. Sleep during naturally occurring respiratory infections: A pilot study. Brain Behav Immun 2019; 79:236-243. [PMID: 30742884 PMCID: PMC7127143 DOI: 10.1016/j.bbi.2019.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 02/06/2023] Open
Abstract
There is strong experimental support that infections increase the drive for sleep in animals, and it is widely believed that more sleep is part of an adaptive immune response. While respiratory infections (RI) are very prevalent in humans, there is a striking lack of systematic knowledge on how it affects sleep. We recruited 100 people, among whom 28 became sick with an RI during the study period (fulfilling criteria for influenza-like illness, ILI, or acute respiratory infection, ARI). We measured sick participants' sleep at home, both objectively (actigraphy) and subjectively (diary ratings), for one week as well as four weeks later when healthy. During the week with RI, people spent objectively longer time in bed and had a longer total sleep time compared to the healthy week. During the infection, participants also had more awakenings, but no significant differences in sleep latency or sleep efficiency. While sick, people also reported increased difficulties falling asleep, worse sleep quality, more restless sleep and more shallow sleep, while they did not report sleep to be less sufficient. Most problems occurred at the beginning of the sickness week, when symptoms were strong, and showed signs of recovery thereafter (as indicated by interactions between condition and day/night of data collection for all the 10 sleep outcomes). The degree of symptoms of RI was related to a worse sleep quality and more restless sleep, but not to any of the objective sleep outcomes or the other subjective sleep variables. Having a higher body temperature was not significantly related to any of the sleep variables. This study suggests that having a respiratory infection is associated with spending more time in bed and sleeping longer, but also with more disturbed sleep, both objectively and subjectively. This novel study should be seen as being of pilot character. There is a need for larger studies which classify pathogen type and include baseline predictors, or that manipulate sleep, in order to understand whether the sleep alterations seen during infections are adaptive and whether sleep interventions could be used to improve recovery from respiratory infections.
Collapse
Affiliation(s)
- Julie Lasselin
- Stress Research Institute, Stockholm University, Stockholm, Sweden,Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Ingre
- Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Regenbogen
- Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, Aachen, Germany,JARA – BRAIN Institute 1: Structure-Function Relationship: Decoding the Human Brain at Systemic Levels, Forschungszentrum Jülich, Jülich, Germany
| | - Mats J. Olsson
- Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Garke
- Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden
| | - Mia Brytting
- Unit for Laboratory Surveillance of Viral Pathogens and Vaccine Preventable Diseases, The Public Health Agency of Sweden, Solna, Sweden
| | - Rachel Edgar
- Molecular Virology, Department of Medicine, Imperial College London, London, UK
| | - Mats Lekander
- Stress Research Institute, Stockholm University, Stockholm, Sweden,Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden
| | - John Axelsson
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Hinojosa-Godinez A, Jave-Suarez LF, Flores-Soto M, Gálvez-Contreras AY, Luquín S, Oregon-Romero E, González-Pérez O, González-Castañeda RE. Melatonin modifies SOX2 + cell proliferation in dentate gyrus and modulates SIRT1 and MECP2 in long-term sleep deprivation. Neural Regen Res 2019; 14:1787-1795. [PMID: 31169197 PMCID: PMC6585545 DOI: 10.4103/1673-5374.257537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Melatonin is a pleiotropic molecule that, after a short-term sleep deprivation, promotes the proliferation of neural stem cells in the adult hippocampus. However, this effect has not been observed in long-term sleep deprivation. The precise mechanism exerted by melatonin on the modulation of neural stem cells is not entirely elucidated, but evidence indicates that epigenetic regulators may be involved in this process. In this study, we investigated the effect of melatonin treatment during a 96-hour sleep deprivation and analyzed the expression of epigenetic modulators predicted by computational text mining and keyword clusterization. Our results showed that the administration of melatonin under sleep-deprived conditions increased the MECP2 expression and reduced the SIRT1 expression in the dentate gyrus. We observed that let-7b, mir-132, and mir-124 were highly expressed in the dentate gyrus after melatonin administration, but they were not modified by sleep deprivation. In addition, we found more Sox2+/5-bromo-2′-deoxyuridine (BrdU)+ cells in the subgranular zone of the sleep-deprived group treated with melatonin than in the untreated group. These findings may support the notion that melatonin modifies the expression of epigenetic mediators that, in turn, regulate the proliferation of neural progenitor cells in the adult dentate gyrus under long-term sleep-deprived conditions. All procedures performed in this study were approved by the Animal Ethics Committee of the University of Guadalajara, Mexico (approval No. CI-16610) on January 2, 2016.
Collapse
Affiliation(s)
- Alan Hinojosa-Godinez
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Luis F Jave-Suarez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, México
| | - Mario Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, México
| | - Alma Y Gálvez-Contreras
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias; Unidad de Atención en Neurociencias, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Sonia Luquín
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas (IICB), Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Oscar González-Pérez
- Laboratorio de Neurociencias, Facultad de Psicología, Universidad de Colima, Colima, México
| | - Rocio E González-Castañeda
- Laboratorio de Microscopía de Alta Resolución, Departamento de Neurociencias; Unidad de Atención en Neurociencias, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| |
Collapse
|
42
|
Pearce SC, Coia HG, Karl JP, Pantoja-Feliciano IG, Zachos NC, Racicot K. Intestinal in vitro and ex vivo Models to Study Host-Microbiome Interactions and Acute Stressors. Front Physiol 2018; 9:1584. [PMID: 30483150 PMCID: PMC6240795 DOI: 10.3389/fphys.2018.01584] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome is extremely important for maintaining homeostasis with host intestinal epithelial, neuronal, and immune cells and this host-microbe interaction is critical during times of stress or disease. Environmental, nutritional, and cognitive stress are just a few factors known to influence the gut microbiota and are thought to induce microbial dysbiosis. Research on this bidirectional relationship as it pertains to health and disease is extensive and rapidly expanding in both in vivo and in vitro/ex vivo models. However, far less work has been devoted to studying effects of host-microbe interactions on acute stressors and performance, the underlying mechanisms, and the modulatory effects of different stressors on both the host and the microbiome. Additionally, the use of in vitro/ex vivo models to study the gut microbiome and human performance has not been researched extensively nor reviewed. Therefore, this review aims to examine current evidence concerning the current status of in vitro and ex vivo host models, the impact of acute stressors on gut physiology/microbiota as well as potential impacts on human performance and how we can parlay this information for DoD relevance as well as the broader scientific community. Models reviewed include widely utilized intestinal cell models from human and animal models that have been applied in the past for stress or microbiology research as well as ex vivo organ/tissue culture models and new innovative models including organ-on-a-chip and co-culture models.
Collapse
Affiliation(s)
- Sarah C Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Heidi G Coia
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, Washington, DC, United States.,711th Human Performance Wing, Airforce Research Laboratory, Airman Systems Directorate, Human-Centered ISR Division, Molecular Mechanisms Branch, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - J P Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Ida G Pantoja-Feliciano
- Soldier Protection and Optimization Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kenneth Racicot
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
43
|
Karl JP, Hatch AM, Arcidiacono SM, Pearce SC, Pantoja-Feliciano IG, Doherty LA, Soares JW. Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota. Front Microbiol 2018; 9:2013. [PMID: 30258412 PMCID: PMC6143810 DOI: 10.3389/fmicb.2018.02013] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Stress, a ubiquitous part of daily human life, has varied biological effects which are increasingly recognized as including modulation of commensal microorganisms residing in the gastrointestinal tract, the gut microbiota. In turn, the gut microbiota influences the host stress response and associated sequelae, thereby implicating the gut microbiota as an important mediator of host health. This narrative review aims to summarize evidence concerning the impact of psychological, environmental, and physical stressors on gut microbiota composition and function. The stressors reviewed include psychological stress, circadian disruption, sleep deprivation, environmental extremes (high altitude, heat, and cold), environmental pathogens, toxicants, pollutants, and noise, physical activity, and diet (nutrient composition and food restriction). Stressors were selected for their direct relevance to military personnel, a population that is commonly exposed to these stressors, often at extremes, and in combination. However, the selected stressors are also common, alone or in combination, in some civilian populations. Evidence from preclinical studies collectively indicates that the reviewed stressors alter the composition, function and metabolic activity of the gut microbiota, but that effects vary across stressors, and can include effects that may be beneficial or detrimental to host health. Translation of these findings to humans is largely lacking at present. This gap precludes concluding with certainty that transient or cumulative exposures to psychological, environmental, and physical stressors have any consistent, meaningful impact on the human gut microbiota. However, provocative preclinical evidence highlights a need for translational research aiming to elucidate the impact of stressors on the human gut microbiota, and how the gut microbiota can be manipulated, for example by using nutrition, to mitigate adverse stress responses.
Collapse
Affiliation(s)
- J. Philip Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Adrienne M. Hatch
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Steven M. Arcidiacono
- Soldier Performance Optimization, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Sarah C. Pearce
- Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Ida G. Pantoja-Feliciano
- Soldier Performance Optimization, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Laurel A. Doherty
- Soldier Performance Optimization, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Jason W. Soares
- Soldier Performance Optimization, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
44
|
Pigarev IN, Pigareva ML. Association of sleep impairments and gastrointestinal disorders in the context of the visceral theory of sleep. J Integr Neurosci 2018; 16:143-156. [PMID: 28891506 DOI: 10.3233/jin-170005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
It was noticed long ago that sleep disorders or interruptions to the normal sleep pattern were associated with various gastrointestinal disorders. We review the studies which established the causal link between these disorders and sleep impairment. However, the mechanism of interactions between the quality of sleep and gastrointestinal pathophysiology remained unclear. Recently, the visceral theory of sleep was formulated. This theory proposes that the same brain structures, and particularly the same cortical sensory areas, which in wakefulness are involved in processing of the exteroceptive information, switch during sleep to the processing of information coming from various visceral systems. We review the studies which demonstrated that neurons of the various cortical areas (occipital, parietal, frontal) during sleep began to fire in response to activation coming from the stomach and small intestine. These data demonstrate that, during sleep, the computational power of the central nervous system, including all cortical areas, is engaged in restoration of visceral systems. Thus, the general mechanism of the interaction between quality of sleep and health became clear.
Collapse
Affiliation(s)
- Ivan N Pigarev
- Institute for Information Transmission Problems (Kharkevich Institute), Russian Academy of Sciences, Bol'shoy Karetniy st. 19, Moscow, 127994, Russia. E-mail:
| | - Marina L Pigareva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova st. 5-a, Moscow, 117485, Russia. E-mail:
| |
Collapse
|
45
|
Interactions of Gut Microbiota, Endotoxemia, Immune Function, and Diet in Exertional Heatstroke. JOURNAL OF SPORTS MEDICINE 2018; 2018:5724575. [PMID: 29850597 PMCID: PMC5926483 DOI: 10.1155/2018/5724575] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022]
Abstract
Exertional heatstroke (EHS) is a medical emergency that cannot be predicted, requires immediate whole-body cooling to reduce elevated internal body temperature, and is influenced by numerous host and environmental factors. Widely accepted predisposing factors (PDF) include prolonged or intense exercise, lack of heat acclimatization, sleep deprivation, dehydration, diet, alcohol abuse, drug use, chronic inflammation, febrile illness, older age, and nonsteroidal anti-inflammatory drug use. The present review links these factors to the human intestinal microbiota (IM) and diet, which previously have not been appreciated as PDF. This review also describes plausible mechanisms by which these PDF lead to EHS: endotoxemia resulting from elevated plasma lipopolysaccharide (i.e., a structural component of the outer membrane of Gram-negative bacteria) and tissue injury from oxygen free radicals. We propose that recognizing the lifestyle and host factors which are influenced by intestine-microbial interactions, and modifying habitual dietary patterns to alter the IM ecosystem, will encourage efficient immune function, optimize the intestinal epithelial barrier, and reduce EHS morbidity and mortality.
Collapse
|
46
|
Pedroni MN, Hirotsu C, Porro AM, Tufik S, Andersen ML. The role of sleep in pemphigus: a review of mechanisms and perspectives. Arch Dermatol Res 2017; 309:659-664. [PMID: 28726005 DOI: 10.1007/s00403-017-1765-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/25/2022]
Abstract
Pemphigus is an autoimmune bullous disease that causes the development of blisters and erosions on the skin and/or mucosa. Its inflammatory process is mediated by cytokines, which interact with sleep in a bidirectional manner. Pain, a frequent symptom due to pemphigus lesions, is well known to impair sleep quality. Depression is also associated with pemphigus and pro-inflammatory cytokines and may impair sleep. Additionally, a common relationship among other dermatological diseases and sleep has increasingly been described. Poor sleep quality is associated with an increased risk for autoimmune diseases, and insomnia is a comorbidity that has recently been associated with pemphigus. Thus, this review will explore the evidence supporting the likely bidirectional relationship between pemphigus and sleep quality and its possible mechanisms involved. This approach covering both pemphigus and sleep will open a research avenue for future studies focusing on the efficacy of the sleep disorders treatment in patients with pemphigus. In the long term, this may provide relevant information to dermatologists regarding new strategies for the management of pemphigus clinical condition, allowing possibly a better quality of life for the patients.
Collapse
Affiliation(s)
- Matheus Negrao Pedroni
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Napoleão de Barros, 925, Vila Clementino, São Paulo, SP, 04024-002, Brazil
| | - Camila Hirotsu
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Napoleão de Barros, 925, Vila Clementino, São Paulo, SP, 04024-002, Brazil
| | - Adriana Maria Porro
- Departamento de Dermatologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Napoleão de Barros, 925, Vila Clementino, São Paulo, SP, 04024-002, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Napoleão de Barros, 925, Vila Clementino, São Paulo, SP, 04024-002, Brazil.
| |
Collapse
|
47
|
Karl JP, Margolis LM, Madslien EH, Murphy NE, Castellani JW, Gundersen Y, Hoke AV, Levangie MW, Kumar R, Chakraborty N, Gautam A, Hammamieh R, Martini S, Montain SJ, Pasiakos SM. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am J Physiol Gastrointest Liver Physiol 2017; 312:G559-G571. [PMID: 28336545 DOI: 10.1152/ajpgi.00066.2017] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 02/08/2023]
Abstract
The magnitude, temporal dynamics, and physiological effects of intestinal microbiome responses to physiological stress are poorly characterized. This study used a systems biology approach and a multiple-stressor military training environment to determine the effects of physiological stress on intestinal microbiota composition and metabolic activity, as well as intestinal permeability (IP). Soldiers (n = 73) were provided three rations per day with or without protein- or carbohydrate-based supplements during a 4-day cross-country ski-march (STRESS). IP was measured before and during STRESS. Blood and stool samples were collected before and after STRESS to measure inflammation, stool microbiota, and stool and plasma global metabolite profiles. IP increased 62 ± 57% (mean ± SD, P < 0.001) during STRESS independent of diet group and was associated with increased inflammation. Intestinal microbiota responses were characterized by increased α-diversity and changes in the relative abundance of >50% of identified genera, including increased abundance of less dominant taxa at the expense of more dominant taxa such as Bacteroides Changes in intestinal microbiota composition were linked to 23% of metabolites that were significantly altered in stool after STRESS. Together, pre-STRESS Actinobacteria relative abundance and changes in serum IL-6 and stool cysteine concentrations accounted for 84% of the variability in the change in IP. Findings demonstrate that a multiple-stressor military training environment induced increases in IP that were associated with alterations in markers of inflammation and with intestinal microbiota composition and metabolism. Associations between IP, the pre-STRESS microbiota, and microbiota metabolites suggest that targeting the intestinal microbiota could provide novel strategies for preserving IP during physiological stress.NEW & NOTEWORTHY Military training, a unique model for studying temporal dynamics of intestinal barrier and intestinal microbiota responses to stress, resulted in increased intestinal permeability concomitant with changes in intestinal microbiota composition and metabolism. Prestress intestinal microbiota composition and changes in fecal concentrations of metabolites linked to the microbiota were associated with increased intestinal permeability. Findings suggest that targeting the intestinal microbiota could provide novel strategies for mitigating increases in intestinal permeability during stress.
Collapse
Affiliation(s)
- J Philip Karl
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts;
| | - Lee M Margolis
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | | | - Nancy E Murphy
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - John W Castellani
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | | | - Allison V Hoke
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland.,Geneva Foundation, Fort Detrick, Maryland; and
| | - Michael W Levangie
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland.,Geneva Foundation, Fort Detrick, Maryland; and
| | - Raina Kumar
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nabarun Chakraborty
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland.,Geneva Foundation, Fort Detrick, Maryland; and
| | - Aarti Gautam
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland
| | - Rasha Hammamieh
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland
| | - Svein Martini
- Norwegian Defense Research Establishment, Kjeller, Norway
| | - Scott J Montain
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Stefan M Pasiakos
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| |
Collapse
|
48
|
Innate immunity modulation in the duodenal mucosa induced by REM sleep deprivation during infection with Trichinella spirallis. Sci Rep 2017; 7:45528. [PMID: 28374797 PMCID: PMC5379483 DOI: 10.1038/srep45528] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Sleep is considered to be an important predictor of the immunity, since the absence of sleep can affect the development of the immune response, and consequently increase the susceptibility to contract an infection. The aim of the present study was to investigate if sleep deprivation and stress induce dysregulation of the duodenal mucous membrane during the acute infection with Trichinella spiralis. Our results shows that, in the intestinal mucous membrane, stress and sleep deprivation, produces different effect in the cells, and this effect depends on the studied duodenal compartment, glands or villi. The sleep deprivation affect mast cells mainly, and the stress response is more heterogeneous. Interestingly, in the duodenal mucous membrane, none population of cells in the infected groups responded equally to both conditions. These findings suggest that the response of the intestinal mucous membrane during the infection caused for T. spiralis turns out to be affected in the sleep-deprived rats, therefore, the results of the present study sustain the theory that sleep is a fundamental process that is capable of modulating the immune response of mucous membranes, particularly the one generated against the parasite Trichinella spiralis.
Collapse
|
49
|
Reynolds AC, Broussard J, Paterson JL, Wright KP, Ferguson SA. Sleepy, circadian disrupted and sick: Could intestinal microbiota play an important role in shift worker health? Mol Metab 2016; 6:12-13. [PMID: 28123932 PMCID: PMC5220389 DOI: 10.1016/j.molmet.2016.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022] Open
Affiliation(s)
- Amy C Reynolds
- Appleton Institute, CQUniversity Australia, 44 Greenhill Road, Wayville, SA 5034, Australia.
| | - Josiane Broussard
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309-0354, USA
| | - Jessica L Paterson
- Appleton Institute, CQUniversity Australia, 44 Greenhill Road, Wayville, SA 5034, Australia
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309-0354, USA
| | - Sally A Ferguson
- Appleton Institute, CQUniversity Australia, 44 Greenhill Road, Wayville, SA 5034, Australia
| |
Collapse
|
50
|
Braga RM, Dourado MN, Araújo WL. Microbial interactions: ecology in a molecular perspective. Braz J Microbiol 2016; 47 Suppl 1:86-98. [PMID: 27825606 PMCID: PMC5156507 DOI: 10.1016/j.bjm.2016.10.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/07/2016] [Indexed: 02/06/2023] Open
Abstract
The microorganism–microorganism or microorganism–host interactions are the key strategy to colonize and establish in a variety of different environments. These interactions involve all ecological aspects, including physiochemical changes, metabolite exchange, metabolite conversion, signaling, chemotaxis and genetic exchange resulting in genotype selection. In addition, the establishment in the environment depends on the species diversity, since high functional redundancy in the microbial community increases the competitive ability of the community, decreasing the possibility of an invader to establish in this environment. Therefore, these associations are the result of a co-evolution process that leads to the adaptation and specialization, allowing the occupation of different niches, by reducing biotic and abiotic stress or exchanging growth factors and signaling. Microbial interactions occur by the transference of molecular and genetic information, and many mechanisms can be involved in this exchange, such as secondary metabolites, siderophores, quorum sensing system, biofilm formation, and cellular transduction signaling, among others. The ultimate unit of interaction is the gene expression of each organism in response to an environmental (biotic or abiotic) stimulus, which is responsible for the production of molecules involved in these interactions. Therefore, in the present review, we focused on some molecular mechanisms involved in the microbial interaction, not only in microbial–host interaction, which has been exploited by other reviews, but also in the molecular strategy used by different microorganisms in the environment that can modulate the establishment and structuration of the microbial community.
Collapse
Affiliation(s)
- Raíssa Mesquita Braga
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Manuella Nóbrega Dourado
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Welington Luiz Araújo
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil.
| |
Collapse
|