1
|
Sivamaruthi BS, Kesika P, Sisubalan N, Chaiyasut C. The Role of Essential Oils on Sleep Quality and Other Sleep-related Issues: Evidence from Clinical Trials. Mini Rev Med Chem 2025; 25:234-258. [PMID: 39225207 DOI: 10.2174/0113895575315700240821054716] [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: 03/11/2024] [Revised: 05/27/2024] [Accepted: 06/07/2024] [Indexed: 09/04/2024]
Abstract
Essential oils (EOs) are a volatile mixture of bioactive compounds extracted from aromatic plants. The composition of EOs varies, which majorly depends on the extraction methods and plant parts. Aromatherapy using EOs has been reported for its several beneficial effects in humans. Aromatherapy is considered a complementary and/ or adjuvant therapeutic approach for treating several illnesses, especially to improve mental health and well-being. The incidence of sleep disorders, specifically insomnia, is nowadays increased, possibly due to urbanization and lifestyle. The studies showed that EOs-based treatments using lavender EO, bergamot EO, cinnamon EO, and rosemary EO (alone or in combinations) could improve sleep quality, duration, and deprivation in healthy subjects and patients, those who suffer from sleep-related issues. The current manuscript details the outcomes of EO-based treatments on the sleep quality of humans and the possible mechanisms associated with the health-promoting properties of EOs. Also, the toxicity and adverse effects of EOs have been discussed. The study indicated that EOs are potent adjuvant therapeutic candidates to manage mood-associated complications in humans. Moreover, the aromatherapeutic field requires detailed studies on toxicity and dose determination, which could provide safe and effective therapeutic results.
Collapse
Affiliation(s)
- Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Faculty of Pharmacy, Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, , Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Periyanaina Kesika
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Faculty of Pharmacy, Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, , Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natarajan Sisubalan
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Faculty of Pharmacy, Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, , Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chaiyavat Chaiyasut
- Faculty of Pharmacy, Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, , Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
2
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
3
|
Zeiher C, Kuhrt H, Rifflet A, Winter K, Boon L, Stassart RM, Nutma E, Middeldorp J, Strating IM, Boneca IG, Bechmann I, Laman JD. Peptidoglycan accumulates in distinct brain regions and cell types over lifetime but is absent in newborns. Brain Behav Immun 2025; 123:799-812. [PMID: 39442638 DOI: 10.1016/j.bbi.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/28/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024] Open
Abstract
Peptidoglycan (PGN) is a large complex polymer critical to structure and function of all bacterial species. Intact PGN and its fragments are inflammatory, contributing to infectious and autoimmune disease. Recent studies show that PGN physiologically contributes to immune setpoints, and importantly also to mouse brain development and behavior. However, for the human brain, it remains unknown whether PGN and its fragments differentially gain access to distinct brain regions, which cell types accumulate it, and whether PGN brain load varies with age. Therefore, we investigated human postmortem brain samples of donors with an extensive age range, from newborns to nonagenarians. We examined two monoclonal antibodies against PGN which were validated using dot blot analysis, competition assays and immunofluorescence experiments on bacteria sacculi, which jointly showed specific detection of Gram-positive PGN. As positive reference tissue, brain tissue from sepsis patients, and human liver were used, both showing the expected high PGN levels. In adult brain tissue of different age (34- to 94-year-old) and sex, we detected PGN signals in seven different brain regions, with highest loads in the occipital cortex, hippocampal formation, frontal cortex, the periventricular region and the olfactory bulb. Age-dependent increase of signals was not evident by microscopic observations and only weak correlation was found by statistical analysis in this cohort. PGN was found intracellularly in the cytoplasm surrounding the cell nucleus in astrocytes, oligodendrocytes, neurons, and endothelial cells, but not in macrophages like microglia. PGN was absent in brain tissues of three human newborns (stillbirth to four weeks old). For comparison, three brain regions from non-human primates of varying age (newborn to 21 years) were immunohistochemically stained. The highest PGN-load was observed in brain tissue from 18- to 21-year-old macaques. This first systematic evaluation of PGN in human postmortem brain suggests that PGN accumulates during lifetime until it reaches a plateau by homeostatic turnover and highlights the ubiquitous presence of PGN in human brain tissues, and their ability to participate in physiological as well as pathological processes throughout life.
Collapse
Affiliation(s)
- Carolin Zeiher
- Institute of Anatomy, University of Leipzig, Leipzig, Germany.
| | - Heidrun Kuhrt
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Aline Rifflet
- Institute Pasteur, Université Paris Cité, INSERM U1306, Biology and Genetics of the Bacterial Cell Wall Unit, F-75015 Paris, France
| | - Karsten Winter
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | | | | | - Erik Nutma
- Biomedical Primate Research Centre (BPRC), Department of Neurobiology and Aging, Rijswijk, Netherlands (The)
| | - Jinte Middeldorp
- Biomedical Primate Research Centre (BPRC), Department of Neurobiology and Aging, Rijswijk, Netherlands (The)
| | - Inge M Strating
- University Groningen, University Medical Center Groningen (UMCG), Dept. Pathology & Medical Biology, and MS Center Noord Nederland (MSCNN), Groningen, Netherlands (The)
| | - Ivo G Boneca
- Institute Pasteur, Université Paris Cité, INSERM U1306, Biology and Genetics of the Bacterial Cell Wall Unit, F-75015 Paris, France.
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany.
| | - Jon D Laman
- University Groningen, University Medical Center Groningen (UMCG), Dept. Pathology & Medical Biology, and MS Center Noord Nederland (MSCNN), Groningen, Netherlands (The).
| |
Collapse
|
4
|
Fang J, Wang S, Liu L, Zhang X, Liu R, Pang X, Cui J, Han J, Zhu X. Gut microbiota: a potential influencer of insomnia occurring after COVID-19 infection. Front Psychiatry 2024; 15:1423715. [PMID: 39109368 PMCID: PMC11300359 DOI: 10.3389/fpsyt.2024.1423715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/09/2024] [Indexed: 04/30/2025] Open
Abstract
The prevalence of insomnia has increased in recent years, significantly affecting the lives of many individuals. Coronavirus disease 2019 (COVID-19) infection has been found to have a substantial impact on the human gut microbiota (GM). Clinical studies have shown that the high prevalence, prolonged duration, and refractory treatment of insomnia symptoms following the COVID-19 pandemic may be related to the effect of COVID-19 infection on the GM. Therefore, the GM may be a potential target for the treatment of insomnia following COVID-19 infection. However, relevant studies have not been well-documented, and the GM has not been sufficiently analyzed in the context of insomnia treatment. Herein, we review the interaction between sleep and the GM, summarize the characteristics of COVID-19-induced abnormal changes in the GM and metabolites in patients with insomnia, and discuss potential mechanisms, including metabolic, immune, and neural pathways, by which these abnormal changes in the GM cause insomnia as well as the factors affecting the GM. Finally, we discuss the prospect of modulating the host GM community for the effective treatment of insomnia after COVID-19 infection and the need for further clinical studies.
Collapse
Affiliation(s)
- Jiale Fang
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siwen Wang
- The Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lijia Liu
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyi Zhang
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruilong Liu
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingchao Pang
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiankun Cui
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jianshu Han
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyu Zhu
- The Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Park JY, Kim MS, Ryu JM. Predictors of sleep quality components in patients undergoing transarterial chemoembolisation: a cross-sectional study. Support Care Cancer 2024; 32:149. [PMID: 38329591 DOI: 10.1007/s00520-024-08338-0] [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: 07/12/2023] [Accepted: 01/20/2024] [Indexed: 02/09/2024]
Abstract
PURPOSE Patients undergoing transarterial chemoembolisation experience postembolisation symptoms and interferences affecting sleep quality, which require intervention. The study aimed to identify the predictors of sleep quality components in patients undergoing transarterial chemoembolisation. METHODS This study included two groups of participants: 50 patients undergoing transarterial chemoembolisation and 45 nurses caring for them. Data were collected from September to November 2022 using a structured questionnaire, and analysed using descriptive statistics, the t-test, analysis of variance, Spearman's rank correlation, and multiple regression analysis using the SPSS 27.0 program (IBM Corp., Armonk, NY, USA). RESULTS The mean sleep quality score was 40.28±14.10. Heat sensation (t=-2.08, p=.043) and fatigue (t=-4.47, p<.001) predicted sleep fragmentation in 38.6% of the patients. Abdominal pain (t=-2.54, p=.014), vomiting (t=-2.21, p=.032), and the expected fatigue by the nurses (t=2.68, p=.014) predicted sleep length in 41.7% of patients. Abdominal pain (t=-2.05, p=.046) explained 42.9% of sleep depth. CONCLUSION Based on the predictors of sleep quality components obtained in this study, strategies to improve sleep quality tailored to patients undergoing transarterial chemoembolisation should be developed. This study highlighted the need to bridge the gap between patients' and nurses' expected fatigue and its contribution to sleep fragmentation and sleep length. It also highlighted the importance of noncontact temperature measurement, controlling vomiting, and pain relief for improving sleep length in patients undergoing transarterial chemoembolisation.
Collapse
Affiliation(s)
- Ji Young Park
- Gastrointestinal Surgery Ward, Pusan National University Hospital, Busan, South Korea
| | - Myoung Soo Kim
- Department of Nursing, Pukyong National University, 599-1, Daeyeon 3 dong, Namgu, Busan, 48513, South Korea.
| | - Jung Mi Ryu
- Department of Nursing, Busan Institute of Science and Technology, Busan, South Korea
| |
Collapse
|
6
|
Hsiao YW, Lin WL, Chou YH, Liu SH, Liao TWE, Chen SA, Lo LW. Renal sympathetic denervation ameliorates the activated inflammatory response through JAK-STAT pathway in a chronic obstructive sleep apnea animal model. Sleep Med 2024; 113:142-151. [PMID: 38016360 DOI: 10.1016/j.sleep.2023.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/05/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVES Obstructive sleep apnea (OSA) is known to increase the risk of cardiovascular disease and inflammation plays a significant role in this process. Renal denervation (RDN) is a novel approach aimed at reducing sympathetic nervous system activity. The role of RDN in the inflammatory response to chronic OSA (COSA) is currently unclear. The main objective was to study inflammatory mechanisms in the rabbit heart with COSA and the effects of RDN. METHODS Eighteen rabbits were randomized into three groups: sham control, COSA, and COSA-RDN. COSA and COSA-RDN groups received liquid silicone injections, while the sham control group received normal saline. We performed combined surgical and chemical RDN through bilateral retroperitoneal flank incisions in the COSA-RDN group after silicone injections. The inflammatory mechanisms were assessed through immunoblotting, real-time PCR, and ELISA after the experiment. RESULTS H&E staining showed immune cell infiltration in COSA, which decreased after RDN treatment. The level of α7nAChR was significantly reduced in COSA compared to the sham control but was restored to a similar level in the COSA-RDN group. Furthermore, the expressions of p-JAK2 and p-STAT3 were significantly reduced in COSA but showed an up-regulation following RDN treatment. Similarly, levels of the inflammatory markers IL-6, IL-1β and TNF-α were markedly increased in COSA but decreased after RDN therapy. We observed NF-κB activation in the COSA rabbit model, which decreased after RDN treatment, as evidenced by decreased NF-κB expression. CONCLUSIONS Our study suggests that RDN treatment may prevent COSA-associated heart inflammation via the JAK2-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ya-Wen Hsiao
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Lun Lin
- Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan; Taichung Veterans General Hospital Taichung, Taiwan
| | - Yu-Hui Chou
- Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Taichung Veterans General Hospital Taichung, Taiwan
| | - Shin-Huei Liu
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Wei Ernie Liao
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Ann Chen
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Taichung Veterans General Hospital Taichung, Taiwan; National Chung Hsing University, Taichung, Taiwan.
| | - Li-Wei Lo
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
7
|
Shirolapov IV, Gribkova OV, Kovalev AM, Shafigullina LR, Ulivanova VA, Kozlov AV, Ereshchenko AA, Lyamin AV, Zakharov AV. [The interactions along the microbiota-gut-brain axis in the regulation of circadian rhythms, sleep mechanisms and disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:79-86. [PMID: 38934670 DOI: 10.17116/jnevro202412405279] [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] [Indexed: 06/28/2024]
Abstract
The bidirectional relationship between cerebral structures and the gastrointestinal tract involving the microbiota embraces the scientific concept of the microbiota-gut-brain axis. The gut microbiome plays an important role in many physiological and biochemical processes of the human body, in the immune response and maintenance of homeostasis, as well as in the regulation of circadian rhythms. There is a relationship between the higher prevalence of a number of neurological disorders, sleep disorders and changes in the intestinal microbiota, which actualizes the study of the complex mechanisms of such correlation for the development of new treatment and prevention strategies. Environmental factors associated with excessive light exposure can aggravate the gut dysbiosis of intestinal microflora, and as a result, lead to sleep disturbances. This review examines the integrative mechanisms of sleep regulation associated with the gut microbiota (the role of neurotransmitters, short-chain fatty acids, unconjugated bile acids, bacterial cell wall components, cytokines). Taking into account the influence of gut dysbiosis as a risk factor in the development of various diseases, the authors systematize key aspects and modern scientific data on the importance of microflora balance to ensure optimal interaction along the microbiota-gut-brain axis in the context of the regulatory role of the sleep-wake cycle and its disorders.
Collapse
Affiliation(s)
| | | | - A M Kovalev
- Samara State Medical University, Samara, Russia
| | | | | | - A V Kozlov
- Samara State Medical University, Samara, Russia
| | | | - A V Lyamin
- Samara State Medical University, Samara, Russia
| | | |
Collapse
|
8
|
Adams GJ, O'Brien PA. The unified theory of sleep: Eukaryotes endosymbiotic relationship with mitochondria and REM the push-back response for awakening. Neurobiol Sleep Circadian Rhythms 2023; 15:100100. [PMID: 37484687 PMCID: PMC10362302 DOI: 10.1016/j.nbscr.2023.100100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
The Unified Theory suggests that sleep is a process that developed in eukaryotic animals from a relationship with an endosymbiotic bacterium. Over evolutionary time the bacterium evolved into the modern mitochondrion that continues to exert an effect on sleep patterns, e.g. the bacterium Wolbachia establishes an endosymbiotic relationship with Drosophila and many other species of insects and is able to change the host's behaviour by making it sleep. The hypothesis is supported by other host-parasite relationships, e.g., Trypanosoma brucei which causes day-time sleepiness and night-time insomnia in humans and cattle. For eukaryotes such as Monocercomonoids that don't contain mitochondria we find no evidence of them sleeping. Mitochondria produce the neurotransmitter gamma aminobutyric acid (GABA), and ornithine a precursor of the neurotransmitter GABA, together with substances such as 3,4dihydroxy phenylalanine (DOPA) a precursor for the neurotransmitter dopamine: These substances have been shown to affect the sleep/wake cycles in animals such as Drosophilia and Hydra. Eukaryote animals have traded the very positive side of having mitochondria providing aerobic respiration for them with the negative side of having to sleep. NREM (Quiet sleep) is the process endosymbionts have imposed upon their host eukaryotes and REM (Active sleep) is the push-back adaptation of eukaryotes with brains, returning to wakefulness.
Collapse
Affiliation(s)
| | - Philip A. O'Brien
- College of Science, Health, Engineering and Education, Murdoch University, WA, Australia
| |
Collapse
|
9
|
Wu J, Zhang B, Zhou S, Huang Z, Xu Y, Lu X, Zheng X, Ouyang D. Associations between gut microbiota and sleep: a two-sample, bidirectional Mendelian randomization study. Front Microbiol 2023; 14:1236847. [PMID: 37645227 PMCID: PMC10461450 DOI: 10.3389/fmicb.2023.1236847] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
Introduction Previous research has reported that the gut microbiota performs an essential role in sleep through the microbiome-gut-brain axis. However, the causal association between gut microbiota and sleep remains undetermined. Methods We performed a two-sample, bidirectional Mendelian randomization (MR) analysis using genome-wide association study summary data of gut microbiota and self-reported sleep traits from the MiBioGen consortium and UK Biobank to investigate causal relationships between 119 bacterial genera and seven sleep-associated traits. We calculated effect estimates by using the inverse-variance weighted (as the main method), maximum likelihood, simple model, weighted model, weighted median, and MR-Egger methods, whereas heterogeneity and pleiotropy were detected and measured by the MR pleiotropy residual sum and outlier method, Cochran's Q statistics, and MR-Egger regression. Results In forward MR analysis, inverse-variance weighted estimates concluded that the genetic forecasts of relative abundance of 42 bacterial genera had causal effects on sleep-associated traits. In the reverse MR analysis, sleep-associated traits had a causal effect on 39 bacterial genera, 13 of which overlapped with the bacterial genera in the forward MR analysis. Discussion In conclusion, our research indicates that gut microbiota may be involved in the regulation of sleep, and conversely, changes in sleep-associated traits may also alter the abundance of gut microbiota. These findings suggest an underlying reciprocal causal association between gut microbiota and sleep.
Collapse
Affiliation(s)
- Jun Wu
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Baofu Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shengjie Zhou
- Department of Obstetrics and Gynecology, Taizhou Women and Children’s Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Ziyi Huang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yindong Xu
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinwu Lu
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangtao Zheng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dong Ouyang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Obstetrics and Gynecology, Taizhou Women and Children’s Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
10
|
Frank MG, Fleshner M, Maier SF. Exploring the immunogenic properties of SARS-CoV-2 structural proteins: PAMP:TLR signaling in the mediation of the neuroinflammatory and neurologic sequelae of COVID-19. Brain Behav Immun 2023; 111:259-269. [PMID: 37116592 PMCID: PMC10132835 DOI: 10.1016/j.bbi.2023.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/07/2023] [Accepted: 04/23/2023] [Indexed: 04/30/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) produces an array of neurologic and neuropsychiatric symptoms in the acute and post-acute phase of infection (PASC; post-acute sequelae of SARS-CoV-2 infection). Neuroinflammatory processes are considered key factors in the etiology of these symptoms. Several mechanisms underpinning the development of inflammatory events in the brain have been proposed including SARS-CoV-2 neurotropism and peripheral inflammatory responses (i.e., cytokine storm) to infection, which might produce neuroinflammation via immune-to-brain signaling pathways. In this review, we explore evidence in support of an alternate mechanism whereby structural proteins (e.g., spike and spike S1 subunit) derived from SARS-CoV-2 virions function as pathogen-associated molecular patterns (PAMPs) to elicit proinflammatory immune responses in the periphery and/or brain via classical Toll-Like Receptor (TLR) inflammatory pathways. We propose that SARS-CoV-2 structural proteins might directly produce inflammatory processes in brain independent of and/or in addition to peripheral proinflammatory effects, which might converge to play a causal role in the development of neurologic/neuropsychiatric symptoms in COVID-19.
Collapse
Affiliation(s)
- Matthew G Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder CO 80301, United States.
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder CO 80301, United States
| | - Steven F Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder CO 80301, United States
| |
Collapse
|
11
|
Sasso J, Ammar RM, Tenchov R, Lemmel S, Kelber O, Grieswelle M, Zhou QA. Gut Microbiome-Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and Disorders. ACS Chem Neurosci 2023; 14:1717-1763. [PMID: 37156006 PMCID: PMC10197139 DOI: 10.1021/acschemneuro.3c00127] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
Gut microbiota includes a vast collection of microorganisms residing within the gastrointestinal tract. It is broadly recognized that the gut and brain are in constant bidirectional communication, of which gut microbiota and its metabolic production are a major component, and form the so-called gut microbiome-brain axis. Disturbances of microbiota homeostasis caused by imbalance in their functional composition and metabolic activities, known as dysbiosis, cause dysregulation of these pathways and trigger changes in the blood-brain barrier permeability, thereby causing pathological malfunctions, including neurological and functional gastrointestinal disorders. In turn, the brain can affect the structure and function of gut microbiota through the autonomic nervous system by regulating gut motility, intestinal transit and secretion, and gut permeability. Here, we examine data from the CAS Content Collection, the largest collection of published scientific information, and analyze the publication landscape of recent research. We review the advances in knowledge related to the human gut microbiome, its complexity and functionality, its communication with the central nervous system, and the effect of the gut microbiome-brain axis on mental and gut health. We discuss correlations between gut microbiota composition and various diseases, specifically gastrointestinal and mental disorders. We also explore gut microbiota metabolites with regard to their impact on the brain and gut function and associated diseases. Finally, we assess clinical applications of gut-microbiota-related substances and metabolites with their development pipelines. We hope this review can serve as a useful resource in understanding the current knowledge on this emerging field in an effort to further solving of the remaining challenges and fulfilling its potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Ramy M. Ammar
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Rumiana Tenchov
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Steven Lemmel
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Olaf Kelber
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Malte Grieswelle
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Qiongqiong Angela Zhou
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| |
Collapse
|
12
|
Feng W, Yang Z, Liu Y, Chen R, Song Z, Pan G, Zhang Y, Guo Z, Ding X, Chen L, Wang Y. Gut microbiota: A new target of traditional Chinese medicine for insomnia. Biomed Pharmacother 2023; 160:114344. [PMID: 36738504 DOI: 10.1016/j.biopha.2023.114344] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
All species have a physiological need for sleep, and sleep is crucial for the preservation and restoration of many physiological processes in the body. Recent research on the effects of gut microbiota on brain function has produced essential data on the relationship between them. It has been discovered that dysregulation of the gut-brain axis is related to insomnia. Certain metabolites of gut microbiota have been linked to insomnia, and disturbances in gut microbiota can worsen insomnia. Traditional Chinese medicine (TCM) has unique advantages for the treatment of insomnia. Taking the gut microbiota as the target and determining the scientific relevance of TCM to the prevention and treatment of insomnia may lead to new concepts for the treatment of sleep disorders and improve the therapeutic effect of sleep. Taking the gut microbiota as an entry point, this paper reviews the relationship between gut microbiota and TCM, the relationship between gut microbiota and insomnia, the mechanism by which gut microbiota regulate sleep, and the mechanism by which TCM regulates gut microbiota for insomnia prevention and treatment. This review provides new ideas for the prevention and treatment of insomnia through TCM and new ideas for drug development.
Collapse
Affiliation(s)
- Wanying Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yangxi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Rui Chen
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihui Song
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Guiyun Pan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yuhang Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zehui Guo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xinya Ding
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
13
|
Sun J, Chen Y, Sun Y, Yang B, Zhou J. Short sleep duration associated with increased risk for new-onset cardiovascular diseases in individuals with metabolic syndromes: Evidence from the China Health and Retirement Longitudinal Study. Front Cardiovasc Med 2022; 9:1010941. [PMID: 36419500 PMCID: PMC9678247 DOI: 10.3389/fcvm.2022.1010941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
To explore the impact and risk of short sleep duration (sleep duration < 6 h/night) on new-onset cardiovascular and cerebrovascular diseases (CVDs) in people with metabolic syndromes (Mets), this study used the 2011 baseline and 2015 follow-up data from the China Longitudinal Study of Health and Retirement (CHARLS) to conduct a prospective study of people aged ≥ 45 years in China. A total of 5,530 individuals without pre-existing CVDs in baseline were included. Mets were defined according to the harmonized criteria. We applied the Logistic Regression (LR), the Deep Neural Networks (DNN), and the Adaptive Boosting (AdaBoost), to evaluate the association between Mets components, short sleep, and the risk of new-onset CVDs, and the importance of multiple variates for new-onset CVDs. During the 4-year follow-up period, 512 individuals developed CVDs, and short sleep increased the risk of CVD in individuals with Mets. The odds ratio for prevalent CVD in Mets with short sleep group was 3.73 (95%CI 2.95–4.71; P < 0.001) compared to the normal group, and 1.99 (95% CI 1.58–2.51; P < 0.001) compared to the Mets without short sleep group. The DNN method reached the highest precision of 92.24% and f1-score of 95.86%, and the Adaboost method reached the highest recall of 99.92%. Both DNN and Adaboost have better predictive performance than LR and revealed short sleep duration and components of Mets are all the strongest predictors of CVD onset.
Collapse
Affiliation(s)
- Jiaxin Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yizhou Chen
- School of Computer Science, Wuhan University, Wuhan, China
| | - Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Bo Yang
| | - Jining Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Jining Zhou
| |
Collapse
|
14
|
Zhanfeng N, Liang W, Jing K, Jinbo B, Yanjun C, Hechun X. Regulation of sleep disorders in patients with traumatic brain injury by intestinal flora based on the background of brain-gut axis. Front Neurosci 2022; 16:934822. [PMID: 36303945 PMCID: PMC9594989 DOI: 10.3389/fnins.2022.934822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/06/2022] [Indexed: 12/01/2023] Open
Abstract
OBJECTIVE This study investigates whether people with sleep disorders following traumatic brain injury exhibit altered intestinal flora. The changes may allow us to gain a better understanding of the role of intestinal flora in patients with sleep disorders after traumatic brain injury, which may give us insights into curing the sleep disorder after traumatic brain injury (TBI). METHOD We analyzed the intestinal microbial colony structure in the feces of the 28 patients in the normal sleep group and the sleep disorder group by 16SrDNAsequencing technology. The bioinformatics method was used to analyze the intestinal flora change in the v3-v4 region of patients with biorhythm disorder and to observe the difference between the two groups. RESULTS Group grouping comparison and analysis of the evolutionary cladistic map showed the intestinal flora of patients with normal sleep after TBI was mainly Bacilli and Lactobacillales, while that of patients with sleep disorders was mainly Lachnospiraceae and Bacteroidales. The histogram of group value distribution by grouping comparison and analysis showed that Lachnospiraceae, Bacteroidales, Bacteroidia, and Bacteroidetes were dominant in the sleep disorder group. A relative abundance map of species with significant differences by group grouping comparison showed the main manifestations of intestinal flora are Firmicutes, Bacilli, Lactobacillales, Streptococcaceae, and Bacteroidetes. The normal sleep group was dominated by Bacilli, Lactobacillales, Streptococcus, and Veillonella, while in the sleep disorder group, Lachnospiraceae, Bacteroidales, Bacteroidia, and Bacteroidetes were the main species. It was found that there were also significant differences in intestinal flora abundance between the two groups after TBI. After statistics processing, it was compared with the normal sleep group, Lactobacillus, Streptococcus, Oribacterium and Rothia, Actinomyces, Streptophyta, TM7-3 bacteria, and Serratia, showing a significant reduction in the sleep disorder group (P < 0.05). However, Odoribacter, Lachnospiraceae, and Bilophila increased significantly (P < 0.05). CONCLUSION The sleep disorders of patients after TBI can be closely related to intestinal flora disturbance, and its internal mechanism needs further study. Intestinal flora has the potential to be a new therapeutic target.
Collapse
Affiliation(s)
- Niu Zhanfeng
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wu Liang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Kang Jing
- ENT & HN Surgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Bai Jinbo
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Chen Yanjun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xia Hechun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
15
|
Lawrence K, Myrissa K, Toribio-Mateas M, Minini L, Gregory AM. Trialling a microbiome-targeted dietary intervention in children with ADHD-the rationale and a non-randomised feasibility study. Pilot Feasibility Stud 2022; 8:108. [PMID: 35606889 PMCID: PMC9125862 DOI: 10.1186/s40814-022-01058-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background Dietary interventions have been previously explored in children with ADHD. Elimination diets and supplementation can produce beneficial behaviour changes, but little is known about the mechanisms mediating change. We propose that these interventions may work, in part, by causing changes in the gut microbiota. A microbiome-targeted dietary intervention was developed, and its feasibility assessed. Methods A non-randomised feasibility study was conducted on nine non-medicated children with ADHD, aged 8–13 years (mean 10.39 years), using a prospective one-group pre-test/post-test design. Participants were recruited from ADHD support groups in London and took part in the 6-week microbiome-targeted dietary intervention, which was specifically designed to impact the composition of gut bacteria. Children were assessed pre- and post-intervention on measures of ADHD symptomatology, cognition, sleep, gut function and stool-sample microbiome analysis. The primary aim was to assess the study completion rate, with secondary aims assessing adherence, adverse events (aiming for no severe and minimal), acceptability and suitability of outcome measures. Results Recruitment proved to be challenging and despite targeting 230 participants directly through support groups, and many more through social media, nine families (of the planned 10) signed up for the trial. The completion rate for the study was excellent at 100%. Exploration of secondary aims revealed that (1) adherence to each aspect of the dietary protocol was very good; (2) two mild adverse events were reported; (3) parents rated the treatment as having good acceptability; (4) data collection and outcome measures were broadly feasible for use in an RCT with a few suggestions recommended; (5) descriptive data for outcome measures is presented and suggests that further exploration of gut microbiota, ADHD symptoms and sleep would be helpful in future research. Conclusions This study provides preliminary evidence for the feasibility of a microbiome-targeted dietary intervention in children with ADHD. Recruitment was challenging, but the diet itself was well-tolerated and adherence was very good. Families wishing to trial this diet may find it an acceptable intervention. However, recruitment, even for this small pilot study, was challenging. Because of the difficulty experienced recruiting participants, future randomised controlled trials may wish to adopt a simpler dietary approach which requires less parental time and engagement, in order to recruit the number of participants required to make meaningful statistical interpretations of efficacy. Trial registration ClinicalTrials.gov Identifier: NCT03737877. Registered 13 November 2018—retrospectively registered, within 2 days of the first participant being recruited. Supplementary Information The online version contains supplementary material available at 10.1186/s40814-022-01058-4.
Collapse
Affiliation(s)
- Kate Lawrence
- Department of Psychology & Pedagogic Science, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK.
| | - Kyriaki Myrissa
- Department of Health Sciences, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK
| | - Miguel Toribio-Mateas
- School of Health and Education, Middlesex University, London, UK.,School of Applied Science, London South Bank University, London, UK
| | - Lori Minini
- Department of Psychology & Pedagogic Science, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK
| | - Alice M Gregory
- Department of Psychology, Goldsmiths, University of London, London, UK
| |
Collapse
|
16
|
Wei Y, Xu J, Miao S, Wei K, Peng L, Wang Y, Wei X. Recent advances in the utilization of tea active ingredients to regulate sleep through neuroendocrine pathway, immune system and intestinal microbiota. Crit Rev Food Sci Nutr 2022; 63:7598-7626. [PMID: 35266837 DOI: 10.1080/10408398.2022.2048291] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sleep disorders have received widespread attention nowadays, which have been promoted by the accelerated pace of life, unhealthy diets and lack of exercise in modern society. The chemical medications to improve sleep has shown serious side effects and risks with high costs. Therefore, it is urgent to develop efficient nutraceuticals from natural sources to ensure sleep quality as a sustainable strategy. As the second most consumed beverage worldwide, the health-promoting effects of tea have long been widely recognized. However, the modulatory effect of teas on sleep disorders has received much less attention. Tea contains various natural sleep-modulating active ingredients such as L-theanine (LTA), caffeine, tea polyphenols (TPP), tea pigments, tea polysaccharides (TPS) and γ-aminobutyric acid (GABA). This review focuses on the potential influence and main regulating mechanisms of different tea active ingredients on sleep, including being absorbed by the small intestine and then cross the blood-brain barrier to act on neurons in the brain as neurotransmitters, manipulating the immune system and further affect sleep-wake cycle by regulating the levels of cytokines, and controlling the gut microbes to maintain the homeostasis of circadian rhythm. Current research progress and limitations are summarized and several future development directions are also proposed. This review hopes to provide new insights into the future elucidation of the sleep-regulating mechanisms of different teas and their natural active ingredients and the development of tea-based functional foods for alleviating sleep disorders. HighlightsNatural sleep-modulating active ingredients in tea have been summarized.Influences of drinking tea or tea active ingredients on sleep are reviewed.Three main regulating mechanisms of tea active ingredients on sleep are explained.The associations among nervous system, immune system and intestinal microbiota are investigated.The potential of developing delivery carriers for tea active ingredients is proposed.
Collapse
Affiliation(s)
- Yang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jia Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Siwei Miao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Kang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lanlan Peng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yuanfeng Wang
- College of Life Sciences, Shanghai Normal University, Shanghai, P.R. China
| | - Xinlin Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
17
|
Lefter R, Cojocariu RO, Ciobica A, Balmus IM, Mavroudis I, Kis A. Interactions between Sleep and Emotions in Humans and Animal Models. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:274. [PMID: 35208598 PMCID: PMC8877042 DOI: 10.3390/medicina58020274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
Recently, increased interest and efforts were observed in describing the possible interaction between sleep and emotions. Human and animal model studies addressed the implication of both sleep patterns and emotional processing in neurophysiology and neuropathology in suggesting a bidirectional interaction intimately modulated by complex mechanisms and factors. In this context, we aimed to discuss recent evidence and possible mechanisms implicated in this interaction, as provided by both human and animal models in studies. In addition, considering the affective component of brain physiological patterns, we aimed to find reasonable evidence in describing the two-way association between comorbid sleep impairments and psychiatric disorders. The main scientific literature databases (PubMed/Medline, Web of Science) were screened with keyword combinations for relevant content taking into consideration only English written papers and the inclusion and exclusion criteria, according to PRISMA guidelines. We found that a strong modulatory interaction between sleep processes and emotional states resides on the activity of several key brain structures, such as the amygdala, prefrontal cortex, hippocampus, and brainstem nuclei. In addition, evidence suggested that physiologically and behaviorally related mechanisms of sleep are intimately interacting with emotional perception and processing which could advise the key role of sleep in the unconscious character of emotional processes. However, further studies are needed to explain and correlate the functional analysis with causative and protective factors of sleep impairments and negative emotional modulation on neurophysiologic processing, mental health, and clinical contexts.
Collapse
Affiliation(s)
- Radu Lefter
- Center of Biomedical Research, Romanian Academy, Iasi Branch, B dul Carol I, no. 8, 700506 Iasi, Romania;
| | - Roxana Oana Cojocariu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, B dul Carol I, no 8, 700505 Iasi, Romania
- Academy of Romanian Scientists, Splaiul Independentei nr. 54, Sector 5, 050094 Bucuresti, Romania
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, Alexandru Lapusneanu Street, no. 26, 700057 Iasi, Romania
| | - Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK;
| | - Anna Kis
- Institute of Cognitive Neuroscience and Psychology, Hungarian Academy of Sciences, 1117 Budapest, Hungary;
| |
Collapse
|
18
|
Wang Z, Yuan K, Ji YB, Li SX, Shi L, Wang Z, Zhou XY, Bao YP, Xie W, Han Y, Shi J, Lu L, Yan W, Chen WH. Alterations of the Gut Microbiota in Response to Total Sleep Deprivation and Recovery Sleep in Rats. Nat Sci Sleep 2022; 14:121-133. [PMID: 35115853 PMCID: PMC8800865 DOI: 10.2147/nss.s334985] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/31/2021] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Accumulating evidence suggests that both sleep loss and gut dysbiosis can lead to metabolic disorders. However, less is known about the impact of total sleep deprivation (SD) and sleep recovery on the composition, function, and metabolic dynamics of the gut microbiota. METHODS Specific-pathogen free Sprague-Dawley rats were subjected to 48 h of SD with gentle handling and then allowed to recover for 1 week. Taxonomic profiles of fecal microbiota were obtained at baseline, 24 h of SD, 48 h of SD, and 1 week of recovery. We used 16S rRNA gene sequencing to analyze the gut microbial composition and function and further characterize microbiota-derived metabolites in rats. RESULTS The microbiota composition analysis revealed that gut microbial composition and metabolites did not change in the rats after 24 h of SD but were significantly altered after 48 h of SD. These changes were reversible after 1 week of sleep recovery. A functional analysis was performed based on Kyoto Encyclopedia of Genes and Genomes (KEGG) annotations, indicating that 19 KEGG pathways were significantly altered in the gut microbiota in SD rats. These functional changes occurred within 24 h of SD, were more apparent after 48 h of SD, and did not fully recover after 1 week of sleep recovery. CONCLUSION These results indicate that acute total SD leads to significant compositional and functional changes in the gut microbiota, and these changes are reversible.
Collapse
Affiliation(s)
- Zhong Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| | - Yan-Bin Ji
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, People’s Republic of China
| | - Su-Xia Li
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, People’s Republic of China
| | - Le Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| | - Zhe Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| | - Xin-Yu Zhou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan-Ping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, People’s Republic of China
| | - Wen Xie
- Mental Health Center of Anhui Province, Hefei, 230032, People’s Republic of China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, People’s Republic of China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, People’s Republic of China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, People’s Republic of China
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, People’s Republic of China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| | - Wen-Hao Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, People’s Republic of China
| |
Collapse
|
19
|
Wang Y, van de Wouw M, Drogos L, Vaghef-Mehrabani E, Reimer RA, Tomfohr-Madsen L, Giesbrecht GF. Sleep and the gut microbiota in preschool-aged children. Sleep 2022; 45:6509073. [PMID: 35037059 PMCID: PMC9189981 DOI: 10.1093/sleep/zsac020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/24/2021] [Indexed: 01/19/2023] Open
Abstract
Sleep plays a significant role in the mental and physical development of children. Emerging evidence in animals and human adults indicates a relationship between sleep and the gut microbiota; however, it is unclear whether the sleep of preschoolers during a key developmental period, associates with features of their gut microbiota. The objective of this study was to assess the relationship between sleep and gut microbiota in preschool-aged children (4.37 ± 0.48 years, n = 143). Sleep measures included total night-time sleep (TST), sleep efficiency (SE), and wake-time after sleep onset (WASO) assessed using actigraphy. Beta-diversity differences between children with low and high TST (p = .048) suggest gut microbiota community differences. Particularly, relative abundance of Bifidobacterium was higher in the high TST group and Bacteroides, was higher in children who had greater SE and less WASO (LDA score >2). In contrast, some Lachnospiraceae members including Blautia and Coprococcus 1 were associated with shorter night-time sleep duration and less efficiency, respectively. We also found a group of fecal metabolites, including specific neuroactive compounds and immunomodulating metabolites were associated with greater sleep efficiency and less time awake at night. Notably, tryptophan and its metabolizing products were higher in children who had higher SE or lower WASO (LDA score >2); concentration of propionate was higher in children with less WASO (p = .036). Overall, our results reveal a novel association between sleep and gut microbiota in preschool-aged children. Longer night-time sleep and greater sleep efficiency were associated with specific commensal bacteria that may regulate sleep through modulating neurotransmitter metabolism and the immune system.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | | | - Lauren Drogos
- Department of Psychology, University of Calgary, Calgary, AB, Canada,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | | | - Raylene A Reimer
- Alberta Children’s Hospital Research Institute (ACHRI), Calgary, AB, Canada,Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lianne Tomfohr-Madsen
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada,Department of Psychology, University of Calgary, Calgary, AB, Canada,Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Gerald F Giesbrecht
- Corresponding author. Gerry Giesbrecht, Department of Psychology, University of Calgary, 2500 University Drive, NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
20
|
Anisman H, Kusnecov AW. Sleep and circadian rhythms. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Li Y, Shao L, Mou Y, Zhang Y, Ping Y. Sleep, circadian rhythm and gut microbiota: alterations in Alzheimer's disease and their potential links in the pathogenesis. Gut Microbes 2021; 13:1957407. [PMID: 34520319 PMCID: PMC8463034 DOI: 10.1080/19490976.2021.1957407] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
ABSTRATCIn recent years, emerging studies have observed gut microbiota (GM) alterations in Alzheimer's disease (AD), even in individuals with mild cognitive impairment (MCI). Further, impaired sleep and circadian patterns are common symptoms of AD, while sleep and circadian rhythm disruption (SCRD) is associated with greater β-amyloid (Aβ) burden and AD risk, sometimes years before the clinical onset of AD. Moreover, reports have demonstrated that GM and its metabolites exhibit diurnal rhythmicity and the role of SCRD in dampening the GM rhythmicity and eubiosis. This review will provide an evaluation of clinical and animal studies describing GM alterations in distinct conditions, including AD, sleep and circadian disruption. It aims to identify the overlapping and distinctive GM alterations in these conditions and their contributions to pathophysiology. Although most studies are observational and use different methodologies, data indicate partial commonalities in GM alterations and unanimity at functional level. Finally, we discuss the possible interactions between SCRD and GM in AD pathogenesis, as well as several methodological improvements that are necessary for future research.
Collapse
Affiliation(s)
- Yi Li
- School of Medicine, Imperial College London, London, UK
| | - Lingzhan Shao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yang Mou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China,Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,CONTACT Yong Ping Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
22
|
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: 0.5] [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
|
23
|
Lapshina KV, Guzhova IV, Ekimova IV. Preventive Administration of the Heat Shock Protein Hsp70 Relieves Endotoxemia-Induced Febrile Reaction in Pigeons ( Columba livia ) and Rats. J EVOL BIOCHEM PHYS+ 2021; 57:1060-1071. [PMID: 34720177 PMCID: PMC8547305 DOI: 10.1134/s0022093021050082] [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: 04/28/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 11/23/2022]
Abstract
The stress-inducible 70 kDa heat shock protein (Hsp70) can
exert a protective effect on endotoxemia and sepsis due to its ability
to interact with immune cells and modulate the immune response.
However, it remains unknown whether Hsp70 is able to relieve endotoxemia-induced fever.
We carried out a comparative study of the effects of preventive
administration of the human recombinant Hsp70 (HSPA1A) on lipopolysaccharide
(LPS)-induced endotoxemia in pigeons and rats with preimplanted
electrodes and thermistors for recording the thermoregulation parameters (brain
temperature, peripheral vasomotor reaction, muscular contractile
activity). Additionally, we analyzed the dynamics of the white blood
cell (WBC) count in rats under the same conditions. It was found
that preventive administration of Hsp70 relieves the LPS-induced
febrile reaction in pigeons and rats and accelerates the restoration
of the WBC count in rats. The data obtained suggest that these warm-blooded
animals share a common physiological mechanism that underlies the
protective effect of Hsp70.
Collapse
Affiliation(s)
- K V Lapshina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - I V Guzhova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - I V Ekimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
24
|
Brenner D, Shorten GD, O'Mahony SM. Postoperative pain and the gut microbiome. NEUROBIOLOGY OF PAIN 2021; 10:100070. [PMID: 34409198 PMCID: PMC8361255 DOI: 10.1016/j.ynpai.2021.100070] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/24/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Poorly controlled postoperative pain remains a major unresolved challenge globally. The gut microbiome impacts on inflammatory pain and neuropathic pain. Microbiota metabolites can regulate peripheral and central sensitisation. Stress is linked to both postoperative pain and an altered gut microbiome.
In excess of 300 million surgical procedures are undertaken worldwide each year. Despite recognition of the prevalence of postoperative pain, and improvements in pain management techniques, poorly controlled postoperative pain remains a major unresolved challenge globally. An estimated 71% and 51% of patients experience moderate to severe pain after surgery in in-patient and outpatient settings, respectively. Inadequately controlled pain after surgery is associated with significant perioperative morbidity including myocardial infarction and pulmonary complications. As many as 20–56% of patients develop chronic pain after commonly performed procedures such as hernia repair, hysterectomy, and thoracotomy. Traditional analgesics and interventions are often ineffective or partially effective in the treatment of postoperative pain, resulting in a chronic pain condition with related socio-economic impacts and reduced quality of life for the patient. Such chronic pain which occurs after surgery is referred to as Persistent Post-Surgical Pain (PPSP). The complex ecosystem that is the gastrointestinal microbiota (including bacteria, fungi, viruses, phage) plays essential roles in the maintenance of the healthy state of the host. A disruption to the balance of this microbiome has been implicated not only in gastrointestinal disease but also neurological disorders including chronic pain. The influence of the gut microbiome is well documented in the context of visceral pain from the gastrointestinal tract while a greater understanding is emerging of the impact on inflammatory pain and neuropathic pain (both of which can occur during the perioperative period). The gut microbiome is an essential source for driving immune maturation and maintaining appropriate immune response. Given that inflammatory processes have been implicated in postoperative pain, aberrant microbiome profiles may play a role in the development of this type of pain. Furthermore, the microorganisms in our gut produce metabolites, neurotransmitters, and neuromodulators which interact with their receptors to regulate peripheral and central sensitisation associated with chronic pain. Microbiota-derived mediators can also regulate neuroinflammation, which is associated with activation of microglia as well as infiltration by immune cells, known to modulate the development and maintenance of central sensitisation. Moreover, risk factors for developing postoperative pain include anxiety, depression, and increased stress response. These central nervous system-related disorders have been associated with an altered gut microbiome and microbiome targeted intervention studies indicate improvements. Females are more likely to suffer from postoperative pain. As gonadal hormones are associated with a differential microbiome and pre-clinical studies show that male microbiome confers protection from inflammatory pain, it is possible that the composition of the microbiome and its by-products contribute to the increased risk for the development of postoperative pain. Very little evidence exists relating the microbiome to somatic pain. Here we discuss the potential role of the gut microbiome in the aetiology and pathophysiology of postoperative pain in the context of other somatic pain syndromes and what is known about microbe-neuron interactions. Investigations are needed to determine the specific role of the gut microbiome in this type of pain which may help inform the development of preventative interventions as well as management strategies to improve patient outcome.
Collapse
Affiliation(s)
- David Brenner
- Department of Anesthesia and Intensive Care Medicine, Cork University Hospital and University College Cork, Ireland
| | - George D Shorten
- Department of Anesthesia and Intensive Care Medicine, Cork University Hospital and University College Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
25
|
Yao C, Wang Z, Jiang H, Yan R, Huang Q, Wang Y, Xie H, Zou Y, Yu Y, Lv L. Ganoderma lucidum promotes sleep through a gut microbiota-dependent and serotonin-involved pathway in mice. Sci Rep 2021; 11:13660. [PMID: 34211003 PMCID: PMC8249598 DOI: 10.1038/s41598-021-92913-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/11/2021] [Indexed: 12/30/2022] Open
Abstract
Ganoderma lucidum is a medicinal mushroom used in traditional Chinese medicine with putative tranquilizing effects. However, the component of G. lucidum that promotes sleep has not been clearly identified. Here, the effect and mechanism of the acidic part of the alcohol extract of G. lucidum mycelia (GLAA) on sleep were studied in mice. Administration of 25, 50 and 100 mg/kg GLAA for 28 days promoted sleep in pentobarbital-treated mice by shortening sleep latency and prolonging sleeping time. GLAA administration increased the levels of the sleep-promoting neurotransmitter 5-hydroxytryptamine and the Tph2, Iptr3 and Gng13 transcripts in the sleep-regulating serotonergic synapse pathway in the hypothalamus during this process. Moreover, GLAA administration reduced lipopolysaccharide and raised peptidoglycan levels in serum. GLAA-enriched gut bacteria and metabolites, including Bifidobacterium, Bifidobacterium animalis, indole-3-carboxylic acid and acetylphosphate were negatively correlated with sleep latency and positively correlated with sleeping time and the hypothalamus 5-hydroxytryptamine concentration. Both the GLAA sleep promotion effect and the altered faecal metabolites correlated with sleep behaviours disappeared after gut microbiota depletion with antibiotics. Our results showed that GLAA promotes sleep through a gut microbiota-dependent and serotonin-associated pathway in mice.
Collapse
Affiliation(s)
- Chunyan Yao
- Key Laboratory of Nutrition of Zhejiang Province, Institute of Health Food, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhiyuan Wang
- Animal Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qianfei Huang
- Key Laboratory of Nutrition of Zhejiang Province, Institute of Health Food, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yin Wang
- Key Laboratory of Nutrition of Zhejiang Province, Institute of Health Food, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hui Xie
- Animal Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Zou
- The Second Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ying Yu
- Key Laboratory of Nutrition of Zhejiang Province, Institute of Health Food, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
26
|
Yan R, Andrew L, Marlow E, Kunaratnam K, Devine A, Dunican IC, Christophersen CT. Dietary Fibre Intervention for Gut Microbiota, Sleep, and Mental Health in Adults with Irritable Bowel Syndrome: A Scoping Review. Nutrients 2021; 13:2159. [PMID: 34201752 PMCID: PMC8308461 DOI: 10.3390/nu13072159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder affecting 4-5% of the global population. This disorder is associated with gut microbiota, diet, sleep, and mental health. This scoping review therefore aims to map existing research that has administrated fibre-related dietary intervention to IBS individuals and reported outcomes on at least two of the three following themes: gut microbiota, sleep, and mental health. Five digital databases were searched to identify and select papers as per the inclusion and exclusion criteria. Five articles were included in the assessment, where none reported on all three themes or the combination of gut microbiota and sleep. Two studies identified alterations in gut microbiota and mental health with fibre supplementation. The other three studies reported on mental health and sleep outcomes using subjective questionnaires. IBS-related research lacks system biology-type studies targeting gut microbiota, sleep, and mental health in patients undergoing diet intervention. Further IBS research is required to explore how human gut microbiota functions (such as short-chain fatty acids) in sleep and mental health, following the implementation of dietary pattern alteration or component supplementation. Additionally, the application of objective sleep assessments is required in order to detect sleep change with more accuracy and less bias.
Collapse
Affiliation(s)
- Ran Yan
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
- Institute for Nutrition Research, Edith Cowan University, Joondalup Drive, Perth 6027, Australia
| | - Lesley Andrew
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
- Institute for Nutrition Research, Edith Cowan University, Joondalup Drive, Perth 6027, Australia
| | - Evania Marlow
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
| | - Kanita Kunaratnam
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
- Institute for Nutrition Research, Edith Cowan University, Joondalup Drive, Perth 6027, Australia
| | - Amanda Devine
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
- Institute for Nutrition Research, Edith Cowan University, Joondalup Drive, Perth 6027, Australia
| | - Ian C. Dunican
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
| | - Claus T. Christophersen
- School of Medical and Health Sciences, Edith Cowan University, Joondalup Drive, Perth 6027, Australia; (L.A.); (E.M.); (K.K.); (A.D.); (I.C.D.); (C.T.C.)
- WA Human Microbiome Collaboration Centre, School of Molecular and Life Sciences, Curtin University, Kent Street, Perth 6102, Australia
- Integrative Metabolomics and Computational Biology Centre, Edith Cowan University, Joondalup Drive, Perth 6027, Australia
| |
Collapse
|
27
|
The Link between Obesity, Microbiota Dysbiosis, and Neurodegenerative Pathogenesis. Diseases 2021; 9:diseases9030045. [PMID: 34201465 PMCID: PMC8293145 DOI: 10.3390/diseases9030045] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
Current research in medicine in several parts of the world has attempted to establish a link between the occurrence of neurodegenerative pathologies, microbiota dysbiosis, and the incidence of obesity. The body’s response to different physicochemical factors has also been influenced by the proper assimilation of bioactive compounds contained in the food that is ingested. Oxidative stress is one of the major factors that directly affects the functioning of the human microbiota. The body’s reaction to this imbalance is crucial to the progression of inflammatory processes, which are based on molecular mechanisms. Microbial dysbiosis can result in a possibly permanent alteration in the physiological response. This review aims to highlight recent contributions made to alleviating human dysbiosis in degenerative diseases, especially for neurodegenerative pathologies based on the rising prevalence of obesity. We discuss the significance of both microbiota modulation and possible alleviations of pathologies by a modulatory function. We argue that pre- and probiotics (including phenolic compounds stimulating the favorable strain from the microbiota) are an effective alternative that can support the microbiota pattern’s modulation over time and the attenuation of indirect causes that determine dysbiosis. Molecular aspects are presented in support of the modulating role of the microbiota following the use of probiotics.
Collapse
|
28
|
Tamm S, Lensmar C, Andreasson A, Axelsson J, Forsberg Morén A, Grunewald J, Gyllfors P, Karshikoff B, Kosek E, Lampa J, Olgart Höglund C, Strand V, Cervenka S, Lekander M. Objective and Subjective Sleep in Rheumatoid Arthritis and Severe Seasonal Allergy: Preliminary Assessments of the Role of Sickness, Central and Peripheral Inflammation. Nat Sci Sleep 2021; 13:775-789. [PMID: 34168509 PMCID: PMC8216747 DOI: 10.2147/nss.s297702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/18/2021] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Disturbed sleep in inflammatory disorders such as allergy and rheumatoid arthritis (RA) is common and may be directly or indirectly related to disease processes, but has not been well characterized in these patient groups, especially not with objective methods. AIM The present study aimed to characterize objective and subjective sleep in patients with allergy or RA using sleep diaries, one-channel EEG and actigraphy. It also aimed to investigate if sleep measures were associated with central immune activation, assessed using translocator protein (TSPO) positron emission tomography, as well as cytokine markers of peripheral inflammation and disease-specific symptoms or general symptoms of sickness. METHODS In total, 18 patients with seasonal pollen allergy, 18 patients with RA and 26 healthy controls were included in the study. Allergy patients and matched controls were assessed twice, in and out of pollen season, and RA patients and controls were assessed once. Sleep was recorded for approximately 1 week at each occasion. RESULTS Patients with allergy had increased levels of slow-wave sleep during pollen season. In contrast, patients with RA had less SWS compared to healthy controls, while no differences were observed in sleep duration or subjective sleep quality. Across groups, neither proinflammatory cytokines, grey matter TSPO levels nor general sickness symptoms were associated with objective or subjective measures of sleep. Rhinitis, but not conjunctivitis, was correlated to worse subjective sleep and more slow wave sleep in allergy. Functional status, but not disease activity, predicted lower subjective sleep in RA. CONCLUSION This study tentatively indicates that both patients with allergy and RA display sleep alterations but does not support inflammation as an independent predictor of the sleep disturbance across these patient groups.
Collapse
Affiliation(s)
- Sandra Tamm
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stress Research Institute, Stockholm University, Stockholm, Sweden
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Catarina Lensmar
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Andreasson
- Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - John Axelsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Anton Forsberg Morén
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services,Region Stockholm, Stockholm, Sweden
| | - Johan Grunewald
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Gyllfors
- Asthma & Allergy Clinic at S:t Görans Hospital, Stockholm, Sweden
| | - Bianka Karshikoff
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jon Lampa
- Department of Medicine, Rheumatology Unit, Center of Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Olgart Höglund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Victoria Strand
- Asthma & Allergy Clinic at S:t Görans Hospital, Stockholm, Sweden
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services,Region Stockholm, Stockholm, Sweden
| | - Mats Lekander
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stress Research Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
29
|
Zhang Y, Cheng L, Liu Y, Wu Z, Weng P. The Intestinal Microbiota Links Tea Polyphenols with the Regulation of Mood and Sleep to Improve Immunity. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1934007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yuting Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Lu Cheng
- Department of Food Science, Rutgers, the State University of New Jersey, New Brunswick, New Jersey, USA
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Peifang Weng
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| |
Collapse
|
30
|
Gyarmati P, Song Y, Dotimas J, Yoshiba G, Christison A. Cross-sectional comparisons of gut microbiome and short-chain fatty acid levels among children with varied weight classifications. Pediatr Obes 2021; 16:e12750. [PMID: 33174684 DOI: 10.1111/ijpo.12750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Limited studies associate changes in microbiota composition and metabolites among children and adolescents with obesity. Decreases in compositional diversity, increases in the proportion of Firmicutes and Bacteroidetes (F/B ratio) and increases in short-chain fatty acids (SCFAs) have been proposed as contributing factors in the pathophysiology of obesity. OBJECTIVES The aim of the current study was to characterize the faecal microbiota composition, diversity, F/B ratio and SCFA levels in different weight categories (lean, overweight, obesity classes 1-3) of children ages 5 to 12 years. METHODS We collected and processed 83 samples from different weight categories (27.7% lean, 11% overweight, 15%, 17% and 17% of obesity classes 1, 2, and 3, respectively). Microbiota content was determined by sequencing the V4 region of the 16S rRNA gene, and SCFA content was analyzed. RESULTS Microbiota compositions showed no significant differences in diversity or F/B ratios between weight categories. However, a relative abundance of Proteobacteria and lack of Verrucomicrobia were demonstrated when comparing severe obesity to the leaner groups. Faecal butyrate, propionate and isopentanoate concentrations increased progressively with weight category demonstrating significance in the class 3 obesity group. CONCLUSIONS Our results show that severe childhood obesity in our study population was associated with changes in gut microbiome composition correlated to previously reported cardiometabolic disease states in obesity. Increased SCFA levels correlate with obesity-related microbiome metabolic function without a reduction in diversity characterized at a phyla level. Further characterization of these specimens at a species level and longitudinal studies are needed to elucidate these relationships.
Collapse
Affiliation(s)
- Peter Gyarmati
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| | - Yajing Song
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| | - James Dotimas
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| | - Grace Yoshiba
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| | - Amy Christison
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA.,Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| |
Collapse
|
31
|
Modulation of Peptidoglycan Synthesis by Recycled Cell Wall Tetrapeptides. Cell Rep 2021; 31:107578. [PMID: 32348759 DOI: 10.1016/j.celrep.2020.107578] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/12/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022] Open
Abstract
The bacterial cell wall is made of peptidoglycan (PG), a polymer that is essential for the maintenance of cell shape and survival. During growth, bacteria remodel their PG, releasing fragments that are predominantly re-internalized and recycled. Here, we show that Vibrio cholerae recycles PG fragments modified with non-canonical d-amino acids (NCDAA), which lead to the accumulation of cytosolic PG tetrapeptides. We demonstrate that the accumulation of recycled tetrapeptides has two regulatory consequences for the cell wall: reduction of d,d-cross-linkage and reduction of PG synthesis. We further demonstrate that l,d-carboxypeptidases from five different species show a preferential activity for substrates containing canonical (d-alanine) versus non-canonical (d-methionine) d-amino acids, suggesting that the accumulation of intracellular tetrapeptides in NCDAA-rich environments is widespread. Collectively, this work reveals a regulatory role of NCDAA linking PG recycling and synthesis to promote optimal cell wall assembly and composition in the stationary phase.
Collapse
|
32
|
Szentirmai É, Massie AR, Kapás L. Lipoteichoic acid, a cell wall component of Gram-positive bacteria, induces sleep and fever and suppresses feeding. Brain Behav Immun 2021; 92:184-192. [PMID: 33307170 PMCID: PMC7897295 DOI: 10.1016/j.bbi.2020.12.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 11/19/2022] Open
Abstract
Fragments of the bacterial cell wall are bioactive microbial molecules that have profound effects on the function of the brain. Some of the cell wall constituents are common to both Gram-positive and Gram-negative bacteria, e.g., peptidoglycans, while other cell wall components are specific to either Gram-positive or Gram-negative microbes. Lipopolysaccharide (LPS), also called endotoxin, is found exclusively in Gram-negative bacteria, while lipoteichoic acid (LTA) is specific to Gram-positive bacteria. The effects of peptidoglycans, their fragments, and LPS are well characterized, they induce sleep, fever and anorexia. In the present study, we investigated the sleep, body temperature and food intake modulating effects of LTA. We found that intraperitoneal injection of 100 and 250 μg LTA from B. subtilis and S. aureus increases non-rapid-eye movement sleep (NREMS) in mice. The effects were dose-dependent, and the changes were accompanied by decreased motor activity and feeding as well as febrile responses. Intraperitoneal injection of 10 μg LTA induced monophasic increases in body temperature, while 100 and 250 μg LTA from B. subtilis induced initial hypothermia followed by fever. Treatment with 250 μg LTA from S. aureus elicited monophasic hypothermia. Administration of 300 μg/kg LTA from S. aureus directly into the portal vein elicited similar sleep responses in rats but did not affect body temperature. The sleep-modulating effects of LTA were similar to that of LPS in mice, although LTA appears to be less potent. These findings suggest that the role of LTA in signaling by Gram-positive bacteria in the host body is analogous to the role of LPS/endotoxin in signaling by Gram-negative microbes. LTA may play a role in the development of sickness response in clinically manifest Gram-positive bacterial infections and may contribute to sleep signaling by the commensal intestinal microbiota.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, United States; Sleep and Performance Research Center, Washington State University, Spokane, WA, United States.
| | - Ashley R Massie
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, United States
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, United States; Sleep and Performance Research Center, Washington State University, Spokane, WA, United States
| |
Collapse
|
33
|
Zhang R, Gao S, Wang S, Zhang J, Bai Y, He S, Zhao P, Zhang H. Gut Microbiota in Patients with Type 1 Narcolepsy. Nat Sci Sleep 2021; 13:2007-2018. [PMID: 34785965 PMCID: PMC8579944 DOI: 10.2147/nss.s330022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/17/2021] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To explore the characteristics of gut microbiota and its relationship between clinical manifestations in patients with type 1 narcolepsy (NT1). PATIENTS AND METHODS Scale and polysomnography were performed in 20 NT1 patients and 16 healthy controls (HC group) to evaluate the clinical characteristics of NT1. Illumina sequencing was performed on bacterial 16S ribosomal RNA gene using V3-V4 regions to compare the fecal microbiota in all subjects. Associations between clinical characteristics and gut microbiota were analyzed using partial correlation analysis. RESULTS Compared with the HC group, the NT1 group had a significantly higher ESS score, longer total sleep time, increased wakefulness, decreased sleep efficiency, disturbance of sleep structure, shorter mean sleep latency, and increased sleep-onset REM periods (all P < 0.05). No differences in alpha and beta diversity were observed between the two groups. In contrast, there were significant differences at the level of class, order, family, and genus (all P < 0.05). LEfSe analysis showed that the relative abundance of Klebsiella in the NT1 group was higher than that in the HC group (P < 0.05), while the relative abundance of Blautia, Barnesiellaceae, Barnesiella, Phocea, Lactococcus, Coriobacteriia, Coriobacteriales, Ruminiclostridium_5, and Bilophila were lower (all P < 0.05). Partial correlation analysis revealed that partial differential bacteria in the NT1 group were correlated with total sleep time, sleep efficiency, stage 1 sleep, arousal index, and sleep latency (all P < 0.05). CONCLUSION Our data revealed differences in intestinal flora structure between NT1 patients and the normal population, thus providing a theoretical basis for future microecological therapy for narcolepsy. However, future larger sample size studies and different study designs are needed to further clarify the possible pathogenesis and potential causality of intestinal flora in NT1 patients and explore the new treatment strategies.
Collapse
Affiliation(s)
- Ruirui Zhang
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China
| | - Shanjun Gao
- Microbiome Laboratory, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Shenghui Wang
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China.,Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yingying Bai
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shuang He
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Pan Zhao
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hongju Zhang
- Department of Neurology, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China.,Department of Neurology, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
34
|
Rijo-Ferreira F, Bjorness TE, Cox KH, Sonneborn A, Greene RW, Takahashi JS. Sleeping Sickness Disrupts the Sleep-Regulating Adenosine System. J Neurosci 2020; 40:9306-9316. [PMID: 33097636 PMCID: PMC7687053 DOI: 10.1523/jneurosci.1046-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/28/2020] [Accepted: 10/11/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with sleeping sickness, caused by the parasite Trypanosoma brucei, have disruptions in both sleep timing and sleep architecture. However, the underlying cause of these sleep disturbances is not well understood. Here, we assessed the sleep architecture of male mice infected with T. brucei and found that infected mice had drastically altered sleep patterns. Interestingly, T. brucei-infected mice also had a reduced homeostatic sleep response to sleep deprivation, a response modulated by the adenosine system. We found that infected mice had a reduced electrophysiological response to an adenosine receptor antagonist and increased adenosine receptor gene expression. Although the mechanism by which T. brucei infection causes these changes remains to be determined, our findings suggest that the symptoms of sleeping sickness may be because of alterations in homeostatic adenosine signaling.SIGNIFICANCE STATEMENT Sleeping sickness is a fatal disease that disrupts the circadian clock, causes disordered temperature regulation, and induces sleep disturbance. To examine the neurologic effects of infection in the absence of other symptoms, in this study, we used a mouse model of sleeping sickness in which the acute infection was treated but brain infection remained. Using this model, we evaluated the effects of the sleeping sickness parasite, Trypanosoma brucei, on sleep patterns in mice, under both normal and sleep-deprived conditions. Our findings suggest that signaling of adenosine, a neuromodulator involved in mediating homeostatic sleep drive, may be reduced in infected mice.
Collapse
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Theresa E Bjorness
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Research Service, VA North Texas Health Care System, Dallas, Texas 75216-7167
| | - Kimberly H Cox
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Alex Sonneborn
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Robert W Greene
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| |
Collapse
|
35
|
Effects of Acupuncture Treatment in Reducing Sleep Disorder and Gut Microbiota Alterations in PCPA-Induced Insomnia Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3626120. [PMID: 33178314 PMCID: PMC7647758 DOI: 10.1155/2020/3626120] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 08/07/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022]
Abstract
Chronic insomnia without intervention will do harm to people's physical and psychological health as well as the quality of life. While ensuring efficacy, traditional Chinese medicine therapy, such as acupuncture, overcomes the side effects of drugs. However, the molecular mechanism of traditional medicine is unclear and it encounters many obstacles in repetitiveness and popularization. On the other side, the placebo effects also need to be eliminated during the intervention. In this study, a number of indicators such as duration of sleep latency, serum markers, pineal gland immunohistochemistry, and gut microbes were detected in the PCPA-induced insomnia mice to compare the effects between acupuncture and hypnotic drug treatments. Although the food intake and weight were not changed, the results show that serum maker and gut microbiota alterations were mediated by concurrent changes in sleep disorder induced by PCPA in mice. Compared with the PCPA-induced insomnia group, dopamine, 5-hydroxytryptamine, and norepinephrine were reduced in serum, and the melatonin was increased in the pineal gland of the acupuncture group as well as zopiclone drug group. Moreover, the analysis results from 16S tag sequencing of the gut microbiome bacterial rRNA hypervariable region show the same improvement effects between the two medical intervention groups. A co-occurrence network analysis showed that blank and acupuncture networks exhibited higher similarity than sham and zopiclone networks and the sham network possessed the highest complexity of microbial communities. Taken together, the gut microbiome will likely be a new target for improving sleep disorders, and taking into account the side effects of hypnotic drugs, nonpharmacological interventions such as acupuncture may be an effective means and have greater clinical benefits.
Collapse
|
36
|
Bacterial Peptidoglycans from Microbiota in Neurodevelopment and Behavior. Trends Mol Med 2020; 26:729-743. [DOI: 10.1016/j.molmed.2020.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023]
|
37
|
Dietary natural cocoa ameliorates disrupted circadian rhythms in locomotor activity and sleep-wake cycles in mice with chronic sleep disorders caused by psychophysiological stress. Nutrition 2020; 75-76:110751. [DOI: 10.1016/j.nut.2020.110751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/16/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022]
|
38
|
Nault S, Creuze V, Al-Omar S, Levasseur A, Nadeau C, Samson N, Imane R, Tremblay S, Carrault G, Pladys P, Praud JP. Cardiorespiratory Alterations in a Newborn Ovine Model of Systemic Inflammation Induced by Lipopolysaccharide Injection. Front Physiol 2020; 11:585. [PMID: 32625107 PMCID: PMC7311791 DOI: 10.3389/fphys.2020.00585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Although it is well known that neonatal sepsis can induce important alterations in cardiorespiratory control, their detailed early features and the mechanisms involved remain poorly understood. As a first step in resolving this issue, the main goal of this study was to characterize these alterations more extensively by setting up a full-term newborn lamb model of systemic inflammation using lipopolysaccharide (LPS) injection. Two 6-h polysomnographic recordings were performed on two consecutive days on eight full-term lambs: the first after an IV saline injection (control condition, CTRL); the second, after an IV injection of 2.5 μg/kg Escherichia coli LPS 0127:B8 (LPS condition). Rectal temperature, locomotor activity, state of alertness, arterial blood gases, respiratory frequency and heart rate, mean arterial blood pressure, apneas and cardiac decelerations, and heart-rate and respiratory-rate variability (HRV and RRV) were assessed. LPS injection decreased locomotor activity (p = 0.03) and active wakefulness (p = 0.01) compared to the CTRL. In addition, LPS injection led to a biphasic increase in rectal temperature (p = 0.01 at ∼30 and 180 min) and in respiratory frequency and heart rate (p = 0.0005 and 0.005, respectively), and to an increase in cardiac decelerations (p = 0.05). An overall decrease in HRV and RRV was also observed. Interestingly, the novel analysis of the representations of the horizontal and vertical visibility network yielded the most statistically significant alterations in HRV structure, suggesting its potential clinical importance for providing an earlier diagnosis of neonatal bacterial sepsis. A second goal was to assess whether the reflexivity of the autonomic nervous system was altered after LPS injection by studying the cardiorespiratory components of the laryngeal and pulmonary chemoreflexes. No difference was found. Lastly, preliminary results provide proof of principle that brainstem inflammation (increased IL-8 and TNF-α mRNA expression) can be shown 6 h after LPS injection. In conclusion, this full-term lamb model of systemic inflammation reproduces several important aspects of neonatal bacterial sepsis and paves the way for studies in preterm lambs aiming to assess both the effect of prematurity and the central neural mechanisms of cardiorespiratory control alterations observed during neonatal sepsis.
Collapse
Affiliation(s)
- Stéphanie Nault
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sally Al-Omar
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Annabelle Levasseur
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Charlène Nadeau
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nathalie Samson
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Roqaya Imane
- CHU Sainte-Justine Research Center, Departments of Neurosciences and Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Sophie Tremblay
- CHU Sainte-Justine Research Center, Departments of Neurosciences and Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Guy Carrault
- Inserm, LTSI - UMR 1099, CHU Rennes, Université Rennes 1, Rennes, France
| | - Patrick Pladys
- Inserm, LTSI - UMR 1099, CHU Rennes, Université Rennes 1, Rennes, France
| | - Jean-Paul Praud
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
39
|
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: 243] [Impact Index Per Article: 48.6] [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
|
40
|
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: 0.4] [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
|
41
|
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: 3.4] [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
|
42
|
Decoeur F, Benmamar-Badel A, Leyrolle Q, Persillet M, Layé S, Nadjar A. Dietary N-3 PUFA deficiency affects sleep-wake activity in basal condition and in response to an inflammatory challenge in mice. Brain Behav Immun 2020; 85:162-169. [PMID: 31100369 DOI: 10.1016/j.bbi.2019.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/12/2019] [Accepted: 05/11/2019] [Indexed: 12/22/2022] Open
Abstract
Essential polyunsaturated fatty acids (PUFA) from the n-3 and n-6 series constitute the building blocks of brain cell membranes where they regulate most aspects of cell physiology. They are either biosynthesized from their dietary precursors or can be directly sourced from the diet. An overall increase in the dietary n-6/n-3 PUFA ratio, as observed in the Western diet, leads to reduced n-3 PUFAs in tissues that include the brain. Some clinical studies have shown a positive correlation between dietary n-3 PUFA intake and sleep quantity, yet evidence is still sparse. We here used a preclinical model of dietary n-3 PUFA deficiency to assess the precise relationship between dietary PUFA intake and sleep/wake activity. Using electroencephalography (EEG)/electromyography (EMG) recordings on n-3 PUFA deficient or sufficient mice, we showed that dietary PUFA deficiency affects the architecture of sleep-wake activity and the oscillatory activity of cortical neurons during sleep. In a second part of the study, and since PUFAs are a potent modulator of inflammation, we assessed the effect of dietary n-3 PUFA deficiency on the sleep response to an inflammatory stimulus known to modulate sleep/wake activity. We injected mice with the endotoxin lipopolysaccharide (LPS) and quantified the sleep response across the following 12 h. Our results revealed that n-3 PUFA deficiency affects the sleep response in basal condition and after a peripheral immune challenge. More studies are now required aimed at deciphering the molecular mechanisms underlying the intimate relationship between n-3 PUFAs and sleep/wake activity.
Collapse
Affiliation(s)
- F Decoeur
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A Benmamar-Badel
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Q Leyrolle
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - M Persillet
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - S Layé
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A Nadjar
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| |
Collapse
|
43
|
Laman JD, 't Hart BA, Power C, Dziarski R. Bacterial Peptidoglycan as a Driver of Chronic Brain Inflammation. Trends Mol Med 2020; 26:670-682. [PMID: 32589935 DOI: 10.1016/j.molmed.2019.11.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022]
Abstract
Peptidoglycan (PGN) is a cell wall component of both Gram-positive and Gram-negative bacteria. Signature fragments of PGN are proinflammatory through engagement of pattern recognition receptors (PRR) on resident tissue cells and circulating leukocytes. Despite its abundance in the gut microbiota, there is limited recognition that PGN could contribute to chronic neuroinflammation. This review highlights current insights into the roles of PGN as a determinant of brain inflammation, notably in multiple sclerosis (MS) and its experimental autoimmune encephalomyelitis (EAE) models. Recent studies demonstrate PGN in blood of healthy adult humans. PGN amplifies autoimmune pathology via activation of innate immune cells. Novel uptake routes through (altered) gut mucosa by myeloid leukocyte subsets promote PGN transport to the brain.
Collapse
Affiliation(s)
- Jon D Laman
- Department of Biomedical Sciences of Cells and Systems, Section of Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Bert A 't Hart
- Department of Biomedical Sciences of Cells and Systems, Section of Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Anatomy and Neuroscience, Free University Amsterdam, Amsterdam, The Netherlands
| | - Christopher Power
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| | - Roman Dziarski
- Indiana University School of Medicine-Northwest, Gary, IN, USA
| |
Collapse
|
44
|
González-Mercado VJ, Sarkar A, Penedo FJ, Pérez-Santiago J, McMillan S, Marrero SJ, Marrero-Falcón MA, Munro CL. Gut microbiota perturbation is associated with acute sleep disturbance among rectal cancer patients. J Sleep Res 2019; 29:e12915. [PMID: 31599472 DOI: 10.1111/jsr.12915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/04/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022]
Abstract
Cancer treatment-associated gut microbial perturbation/dysbiosis has been implicated in the pathobiology of sleep disturbance; however, evidence is scarce. Eighteen newly diagnosed rectal cancer patients (ages 52-81 years; 10 males) completed a sleep disturbance questionnaire and provided stool samples for 16s RNA gene sequencing during chemo-radiotherapy. Descriptive statistics, Wilcoxon test and regression analyses were computed. Regression analyses showed the Shannon's diversity index to be a significant factor associated with sleep disturbance. This preliminary work suggests that the biological "gut-brain axis" mechanism may be associated with symptoms of sleep disturbance.
Collapse
Affiliation(s)
| | - Anujit Sarkar
- College of Nursing, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Frank J Penedo
- Department of Psychology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Susan McMillan
- College of Nursing, University of South Florida, Tampa, FL, USA
| | - Sara Janet Marrero
- College of Art and Sciences, University of South Florida, Tampa, FL, USA
| | | | - Cindy L Munro
- University of Miami School of Nursing and Health Studies, Miami, FL, USA
| |
Collapse
|
45
|
Tosoni G, Conti M, Diaz Heijtz R. Bacterial peptidoglycans as novel signaling molecules from microbiota to brain. Curr Opin Pharmacol 2019; 48:107-113. [PMID: 31557694 DOI: 10.1016/j.coph.2019.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022]
Abstract
Mounting evidence indicates that gut microbiota exerts a broad range of effects on host physiology and development beyond the gastrointestinal tract, including the modulation of brain development. However, the mechanisms mediating the interactions between the microbiota and the developing brain are still poorly understood. Pattern recognition receptors of the innate immune system that recognize microbial products, such as peptidoglycans have emerged as potential key regulators of gut microbiome-brain interactions. Peptidoglycan-sensing molecules are expressed in the placenta and brain during specific time windows of development. Moreover, peptidoglycans are ubiquitously present in circulation and can cross the blood brain barrier. This review brings together the current evidence supporting a broad function of peptidoglycans well beyond host's immunity, extending to neurodevelopment and behavior.
Collapse
Affiliation(s)
- Giorgia Tosoni
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Rochellys Diaz Heijtz
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden; INSERM U1239, University of Rouen Normandy 76130 Mont-Saint-Aignan, France.
| |
Collapse
|
46
|
No changes in gut microbiota after two-week sleep extension in chronically sleep-deprived individuals. Sleep Med 2019; 68:27-30. [PMID: 32018189 DOI: 10.1016/j.sleep.2019.08.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gut microbiota has been linked to obesity and glucose metabolism. Insufficient sleep is also known to be associated with insulin resistance, and sleep extension was reported to improve glucose metabolism in short sleepers. This study aimed to explore whether sleep extension was associated with changes in gut microbiota and whether there was a relationship with glucose parameters. METHODS We performed a secondary analysis of eight short-seeping but otherwise healthy subjects who participated in a cross over study of two-week home sleep extension and two weeks of habitual sleep. After each sleep condition, stool samples were collected and glucose parameters were obtained. Stool DNA extraction was performed and 16S rRNA was sequenced by MiSeq™. The resulting sequence data were processed to infer relative abundances of taxa present and then analyzed to detect any differences in the abundances of the taxa or overall diversity of the microbiome. RESULTS Mean (SD) sleep duration during habitual sleep and sleep extension was 5.58 (0.53) and 6.60 (0.43) hours/night, respectively. Using the Bray-Curtis index, there was no significant dissimilarity of the genus-level microbial community between the two sleeping conditions (ADONIS, R2 = 0.017, p = 0.988 and ANOSIM, R = -0.131, p = 0.991). Within-sample microbial diversity (ie, the Shannon index) also did not find significant differences (p = 0.861). There was no significant relationship between per-individual dissimilarity and objective and subjective sleep variables, or glycemic parameters. Only higher sleep efficiency was related to higher abundance of the phyla Tenericutes. CONCLUSION Two-week sleep extension in short sleepers was not associated with changes in gut microbiota.
Collapse
|
47
|
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: 768] [Impact Index Per Article: 128.0] [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
|
48
|
Szentirmai É, Millican NS, Massie AR, Kapás L. Butyrate, a metabolite of intestinal bacteria, enhances sleep. Sci Rep 2019; 9:7035. [PMID: 31065013 PMCID: PMC6504874 DOI: 10.1038/s41598-019-43502-1] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/25/2019] [Indexed: 12/17/2022] Open
Abstract
Emerging evidence suggests that the intestinal microbiota is a source of sleep-promoting signals. Bacterial metabolites and components of the bacterial cell wall are likely to provide important links between the intestinal commensal flora and sleep-generating mechanisms in the brain. Butyrate is a short-chain fatty acid produced by the intestinal bacteria by the fermentation of nondigestible polysaccharides. We tested the hypothesis that butyrate may serve as a bacterial-derived sleep-promoting signal. Oral gavage administration of tributyrin, a butyrate pro-drug, elicited an almost 50% increase in non-rapid-eye movement sleep (NREMS) in mice for 4 hours after the treatment. Similarly, intraportal injection of butyrate led to prompt and robust increases in NREMS in rats. In the first 6 hours after the butyrate injection, NREMS increased by 70%. Both the oral and intraportal administration of butyrate led to a significant drop in body temperature. Systemic subcutaneous or intraperitoneal injection of butyrate did not have any significant effect on sleep or body temperature. The results suggest that the sleep-inducing effects of butyrate are mediated by a sensory mechanism located in the liver and/or in the portal vein wall. Hepatoportal butyrate-sensitive mechanisms may play a role in sleep modulation by the intestinal microbiota.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America.
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America.
| | - Nicklaus S Millican
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
| | - Ashley R Massie
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| |
Collapse
|
49
|
Huangfu YR, Peng W, Guo BJ, Shen ZF, Li L, Liu SW, Zheng H, Hu YP. Effects of acupuncture in treating insomnia due to spleen-stomach disharmony syndrome and its influence on intestinal microbiome: Study protocol for a randomized controlled trial. JOURNAL OF INTEGRATIVE MEDICINE 2019; 17:161-166. [PMID: 30819614 DOI: 10.1016/j.joim.2019.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 11/21/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND Insomnia is a common complaint that is closely related to gastrointestinal symptoms, which is consistent with the traditional Chinese medicine classical theory of "stomach disharmony leading to restless sleep." Acupuncture is an effective complementary and alternative medicine therapy to improve gastrointestinal function and restore the normal sleep-wake cycle. However, studies on the effectiveness of acupuncture for insomnia due to spleen-stomach disharmony syndrome are limited to case reports and few randomized controlled trials; deeper research on its mechanism is still lacking. This randomized controlled trial aims to assess the treatment efficacy of "harmonizing stomach to tranquilize mind" acupuncture for insomnia and its influence on the intestinal microbiome. METHODS/DESIGN This is a randomized, single-blind, parallel-group study. Sixty eligible patients with insomnia due to spleen-stomach disharmony syndrome will be randomly divided into two groups (1:1 allocation ratio). The intervention group will use "harmonizing stomach to tranquilize mind" acupuncture, and the control group will receive sham acupuncture. Participants will receive 5 acupuncture treatment sessions per week for 4 consecutive weeks. The Pittsburgh Sleep Quality Index will be used to evaluate the clinical efficacy of acupuncture treatment by making assessments at baseline, the end of treatment and the end of the follow-up. High-throughput 16S ribosomal ribonucleic acid gene sequencing will be performed to detect changes in the intestinal microbial composition before and after treatment. DISCUSSION The results of this trial are expected to confirm that "harmonizing stomach to tranquilize mind" acupuncture can effectively relieve insomnia and alter the intestinal microbiome. TRIAL REGISTRATION Chinese Clinical Trials Registry: ChiCTR1800017092.
Collapse
Affiliation(s)
- Ya-Ru Huangfu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Wei Peng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Bao-Jun Guo
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Zhi-Fu Shen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Li Li
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Shi-Wei Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Hui Zheng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - You-Ping Hu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China.
| |
Collapse
|
50
|
Irazoki O, Hernandez SB, Cava F. Peptidoglycan Muropeptides: Release, Perception, and Functions as Signaling Molecules. Front Microbiol 2019; 10:500. [PMID: 30984120 PMCID: PMC6448482 DOI: 10.3389/fmicb.2019.00500] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Peptidoglycan (PG) is an essential molecule for the survival of bacteria, and thus, its biosynthesis and remodeling have always been in the spotlight when it comes to the development of antibiotics. The peptidoglycan polymer provides a protective function in bacteria, but at the same time is continuously subjected to editing activities that in some cases lead to the release of peptidoglycan fragments (i.e., muropeptides) to the environment. Several soluble muropeptides have been reported to work as signaling molecules. In this review, we summarize the mechanisms involved in muropeptide release (PG breakdown and PG recycling) and describe the known PG-receptor proteins responsible for PG sensing. Furthermore, we overview the role of muropeptides as signaling molecules, focusing on the microbial responses and their functions in the host beyond their immunostimulatory activity.
Collapse
Affiliation(s)
| | | | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|