1
|
Bongiovanni T, Santiago M, Zielinska K, Scheiman J, Barsa C, Jäger R, Pinto D, Rinaldi F, Giuliani G, Senatore T, Kostic AD. A Lactobacillus consortium provides insights into the sleep-exercise-microbiome nexus in proof of concept studies of elite athletes and in the general population. MICROBIOME 2025; 13:1. [PMID: 39748236 PMCID: PMC11697739 DOI: 10.1186/s40168-024-01936-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND The complex relationship among sleep, exercise, and the gut microbiome presents a unique opportunity to improve health and wellness. Here, we conducted the first large-scale investigation into the influence of a novel elite athlete-derived probiotic, consisting of a multi-strain Lactobacillus consortium, on sleep quality, exercise recovery, and gut microbiome composition in both elite athletes (n = 11) and the general population (n = 257). RESULTS Our two-phase study design, which included an open-label study followed by a controlled longitudinal study in a professional soccer team, allowed us to identify key interactions between probiotics, the gut microbiome, and the host. In the placebo-controlled study, we observed significant improvements in self-reported sleep quality by 69%, energy levels by 31%, and bowel movements by 37% after probiotic intervention relative to after placebo. These improvements were associated with a significant decrease in D-ROMS (a marker of oxidative stress) and a significantly higher free-testosterone/cortisol ratio. Multi-omics analyses revealed specific changes in microbiome composition and function, potentially providing mechanistic insights into these observed effects. CONCLUSION This study provides novel insights into how a multi-strain Lactobacillus probiotic modulates sleep quality, exercise recovery, and gut microbiome composition in both the general population and elite athletes, and introduces potential mechanisms through which this probiotic could be influencing overall health. Our results emphasize the untapped potential of tailored probiotic interventions derived from extremely fit and healthy individuals in improving several aspects of health and performance directly in humans. Video Abstract.
Collapse
Affiliation(s)
- Tindaro Bongiovanni
- Player Health and Performance, Palermo Football Club, Palermo, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | - Tullio Senatore
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | |
Collapse
|
2
|
Wang Z, Wang Z, Lu T, Yuan G, Chen W, Jin J, Jiang X, Yan W, Yuan K, Zou G, Bao Y, Shi J, Liu X, Wei H, Han Y, Lu L. Gut microbiota regulate insomnia-like behaviors via gut-brain metabolic axis. Mol Psychiatry 2024:10.1038/s41380-024-02867-0. [PMID: 39658705 DOI: 10.1038/s41380-024-02867-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Sleep interacts reciprocally with the gut microbiota. However, mechanisms of the gut microbe-brain metabolic axis that are responsible for sleep behavior have remained largely unknown. Here, we showed that the absence of the gut microbiota can alter sleep behavior. Sleep deprivation reduced butyrate levels in fecal content and the hypothalamus in specific pathogen-free mice but not in germ-free mice. The microbial metabolite butyrate can promote sleep by modulating orexin neuronal activity in the lateral hypothalamic area in mice. Insomnia patients had lower serum butyrate levels and a deficiency in butyrate-producing species within the gut microbiota. Transplantation of the gut microbiota from insomnia patients to germ-free mice conferred insomnia-like behaviors, accompanied by a decrease in serum butyrate levels. The oral administration of butyrate rescued sleep disturbances in recipient mice. Overall, these findings reveal the causal role of microbial metabolic pathways in modulating insomnia-like behaviors, suggesting potential therapeutic strategies for treating sleep disorders.
Collapse
Affiliation(s)
- 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), 100191, Beijing, China
| | - 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), 100191, Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Guohao Yuan
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenhao Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Jin
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400030, 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), 100191, Beijing, 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), 100191, Beijing, China
| | - Guichang Zou
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Xiaoxing Liu
- 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), 100191, Beijing, China.
| | - Hong Wei
- Yu-Yue Pathology Scientific Research Center, Chongqing, 401329, China.
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, 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), 100191, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorders, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
3
|
Li C, Chen S, Wang Y, Su Q. Microbiome-Based Therapeutics for Insomnia. Int J Mol Sci 2024; 25:13208. [PMID: 39684918 DOI: 10.3390/ijms252313208] [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: 10/25/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Insomnia poses considerable risks to both physical and mental health, leading to cognitive impairment, weakened immune function, metabolic dysfunction, cardiovascular issues, and reduced quality of life. Given the significant global increase in insomnia and the growing scientific evidence connecting gut microbiota to this disorder, targeting gut microbiota as an intervention for insomnia has gained popularity. In this review, we summarize current microbiome-based therapeutics for insomnia, including dietary modifications; probiotic, prebiotic, postbiotic, and synbiotic interventions; and fecal microbiota transplantation. Moreover, we assess the capabilities and weaknesses of these technologies to offer valuable insights for future studies.
Collapse
Affiliation(s)
- Chenyu Li
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sizhe Chen
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yun Wang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Wang Y, Pan L, Guan R. Mechanism of Insomnia After Stroke Based on Intestinal Flora. Int J Gen Med 2024; 17:5493-5502. [PMID: 39628982 PMCID: PMC11611988 DOI: 10.2147/ijgm.s488714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/17/2024] [Indexed: 12/06/2024] Open
Abstract
Stroke has emerged as the second leading cause of mortality. Insomnia after stroke is a highly prevalent complication of stroke with a complex mechanism, impacting daily activities and hindering neurological function rehabilitation while also increasing the risk of stroke recurrence. With the development of molecular biology, intestinal flora has garnered considerable interest in the past few years because of its significant implications for human physiology and pathology. Numerous studies have emphasized the crucial function of intestinal flora in the pathological changes associated with insomnia after stroke. It can influence sleep patterns following a stroke by modulating various pathways, including the hypothalamic-pituitary-adrenal (HPA) axis, immune responses, and neural mechanisms. Disruption of intestinal flora can adversely affect post-stroke sleep quality, while sleep after stroke can also lead to intestinal flora imbalance. Based on the intestinal flora, this paper explores the involvement of hypothalamic-pituitary-adrenal axis (HPA axis), immune pathway and neural pathway in insomnia after stroke, aiming to offer insights for the prevention, treatment, and research of post-stroke insomnia.
Collapse
Affiliation(s)
- Yibo Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Limin Pan
- Out-Patient Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Ruiqian Guan
- Massage Department, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
5
|
Szentirmai É, Buckley K, Massie AR, Kapás L. Lipopolysaccharide-mediated effects of the microbiota on sleep and body temperature. Sci Rep 2024; 14:27378. [PMID: 39521828 PMCID: PMC11550806 DOI: 10.1038/s41598-024-78431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Recent research suggests that microbial molecules translocated from the intestinal lumen into the host's internal environment may play a role in various physiological functions, including sleep. Previously, we identified that butyrate, a short-chain fatty acid produced by intestinal bacteria, and lipoteichoic acid, a cell wall component of gram-positive bacteria, induce sleep when their naturally occurring translocation is mimicked by direct delivery into the portal vein. Building upon these findings, we aimed to explore the sleep signaling potential of intraportally administered lipopolysaccharide (LPS), a primary component of gram-negative bacterial cell walls, in rats. Low dose of LPS (1 μg/kg) increased sleep duration and prolonged fever, without affecting systemic LPS levels. Interestingly, administering LPS systemically outside the portal region at a dose 20 times higher did not affect sleep, indicating a localized sensitivity within the hepatoportal region for the sleep and febrile effects of LPS. Furthermore, both the sleep- and fever-inducing effects of LPS were inhibited by indomethacin, a prostaglandin synthesis inhibitor, and replicated by intraportal administration of prostaglandin E2 or arachidonic acid, suggesting the involvement of the prostaglandin system in mediating these actions.
Collapse
Affiliation(s)
- Éva Szentirmai
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA.
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, USA.
| | - Katelin Buckley
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
| | - Ashley R Massie
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
| | - Levente Kapás
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, USA
| |
Collapse
|
6
|
Trouki C, Campanella B, Onor M, Vornoli A, Pozzo L, Longo V, Bramanti E. Probing the alterations in mice cecal content due to high-fat diet. Food Chem 2024; 455:139856. [PMID: 38823144 DOI: 10.1016/j.foodchem.2024.139856] [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: 01/26/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
The global prevalence of obesity more than doubled between 1990 and 2022. By 2022, 2.5 billion adults aged 18 and older were overweight, with over 890 million of them living with obesity. The urgent need for understanding the impact of high-fat diet, together with the demanding of analytical methods with low energy/chemicals consumption, can be fulfilled by rapid, high-throughput spectroscopic techniques. To understand the impact of high-fat diet on the metabolic signatures of mouse cecal contents, we characterized metabolite variations in two diet-groups (standard vs high-fat diet) using FTIR spectroscopy and multivariate analysis. Their cecal content showed distinct spectral features corresponding to high- and low-molecular-weight metabolites. Further quantification of 13 low-molecular-weight metabolites using liquid chromatography showed significant reduction in the production of short chain fatty acids and amino acids associated with high-fat diet samples. These findings demonstrated the potential of spectroscopy to follow changes in gut metabolites.
Collapse
Affiliation(s)
- Cheherazade Trouki
- CNR-IPCF, Institute of Chemical and Physical Processes, National Research Council, via Moruzzi 1, Pisa 56124, Italy; Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Beatrice Campanella
- CNR-ICCOM, Institute of Chemistry of Organometallic Compounds, National Research Council, via Moruzzi 1, Pisa 56124, Italy.
| | - Massimo Onor
- CNR-ICCOM, Institute of Chemistry of Organometallic Compounds, National Research Council, via Moruzzi 1, Pisa 56124, Italy
| | - Andrea Vornoli
- CNR-IBBA, Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, Pisa 56124, Italy
| | - Luisa Pozzo
- CNR-IBBA, Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, Pisa 56124, Italy
| | - Vincenzo Longo
- CNR-IBBA, Institute of Agricultural Biology and Biotechnology, National Research Council, Via Moruzzi 1, Pisa 56124, Italy
| | - Emilia Bramanti
- CNR-ICCOM, Institute of Chemistry of Organometallic Compounds, National Research Council, via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
7
|
Li X, Zhang Y, Zhang Q, Cao A, Feng J. Eucalyptus essential oil exerted a sedative-hypnotic effect by influencing brain neurotransmitters and gut microbes via the gut microbiota-brain axis. Front Pharmacol 2024; 15:1464654. [PMID: 39386024 PMCID: PMC11461282 DOI: 10.3389/fphar.2024.1464654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Sleep disorders are becoming more and more common, leading to many health problems. However, most of current available medications to treat sleep disorders are addictive and even impair cognitive abilities. Therefore, it is important to find a natural and safe alternative to treat sleep disorders. In this study, twenty-four 8-week-old male ICR mice (25 ± 2 g) were equally divided into three groups: the control group (gavage of 0.9% saline), the eucalyptus essential oil (EEO) group (10 mg/kg B.W.), and the diazepam group (1 mg/kg B.W.). Firstly, open field test and sleep induction test were used to determine the sedative-hypnotic effect of EEO. Secondly, the effect of EEO on neurotransmitters in the mice brain was determined. Finally, based on the gut microbiota-brain axis (GMBA), the effect of EEO on the intestinal flora of mice was explored. It was found that EEO significantly reduce the activity and prolong the sleep duration of mice, exhibiting a good sedative-hypnotic effect. In the brain, EEO could increase the levels of sleep-promoting neurotransmitters, such as glutamine, Gamma-aminobutyric acid (GABA), glycine, tryptophan, N-acetylserotonin, and 5-hydroxyindoleacetic acid (5-HIAA). In the intestine, EEO was found to increase the diversity of gut microbes, the abundance of short chain fatty acid (SCFA) producing flora, and the abundance of functional flora synthesizing GABA and glycine neurotransmitters. These studies suggested that EEO exerted a sedative-hypnotic effect by acting on gut microbes and neurotransmitters in the brain. EEO has the potential to become a natural and safe alternative to traditional hypnotic sedative drugs.
Collapse
Affiliation(s)
- Xuejiao Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yuanyi Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qian Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Aizhi Cao
- Biotechnology R&D Center of Shandong Longchang Animal Health Products Co., Ltd., Jinan, China
| | - Jie Feng
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Sharon O, Ben Simon E, Shah VD, Desel T, Walker MP. The new science of sleep: From cells to large-scale societies. PLoS Biol 2024; 22:e3002684. [PMID: 38976664 PMCID: PMC11230563 DOI: 10.1371/journal.pbio.3002684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
In the past 20 years, more remarkable revelations about sleep and its varied functions have arguably been made than in the previous 200. Building on this swell of recent findings, this essay provides a broad sampling of selected research highlights across genetic, molecular, cellular, and physiological systems within the body, networks within the brain, and large-scale social dynamics. Based on this raft of exciting new discoveries, we have come to realize that sleep, in this moment of its evolution, is very much polyfunctional (rather than monofunctional), yet polyfunctional for reasons we had never previously considered. Moreover, these new polyfunctional insights powerfully reaffirm sleep as a critical biological, and thus health-sustaining, requisite. Indeed, perhaps the only thing more impressive than the unanticipated nature of these newly emerging sleep functions is their striking divergence, from operations of molecular mechanisms inside cells to entire group societal dynamics.
Collapse
Affiliation(s)
- Omer Sharon
- Department of Psychology, University of California, Berkeley, California, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Eti Ben Simon
- Department of Psychology, University of California, Berkeley, California, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Vyoma D. Shah
- Department of Psychology, University of California, Berkeley, California, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Tenzin Desel
- Department of Psychology, University of California, Berkeley, California, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Matthew P. Walker
- Department of Psychology, University of California, Berkeley, California, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| |
Collapse
|
9
|
Zhai M, Song W, Liu Z, Cai W, Lin GN. Causality Investigation between Gut Microbiome and Sleep-Related Traits: A Bidirectional Two-Sample Mendelian Randomization Study. Genes (Basel) 2024; 15:769. [PMID: 38927705 PMCID: PMC11202894 DOI: 10.3390/genes15060769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Recent research has highlighted associations between sleep and microbial taxa and pathways. However, the causal effect of these associations remains unknown. To investigate this, we performed a bidirectional two-sample Mendelian randomization (MR) analysis using summary statistics of genome-wide association studies (GWAS) from 412 gut microbiome traits (N = 7738) and GWAS studies from seven sleep-associated traits (N = 345,552 to 386,577). We employed multiple MR methods to assess causality, with Inverse Variance Weighted (IVW) as the primary method, alongside a Bonferroni correction ((p < 2.4 × 10-4) to determine significant causal associations. We further applied Cochran's Q statistical analysis, MR-Egger intercept, and Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) for heterogeneity and pleiotropy assessment. IVW estimates revealed 79 potential causal effects of microbial taxa and pathways on sleep-related traits and 45 inverse causal relationships, with over half related to pathways, emphasizing their significance. The results revealed two significant causal associations: genetically determined relative abundance of pentose phosphate decreased sleep duration (p = 9.00 × 10-5), and genetically determined increase in fatty acid level increased the ease of getting up in the morning (p = 8.06 × 10-5). Sensitivity analyses, including heterogeneity and pleiotropy tests, as well as a leave-one-out analysis of single nucleotide polymorphisms, confirmed the robustness of these relationships. This study explores the potential causal relationships between sleep and microbial taxa and pathways, offering novel insights into their complex interplay.
Collapse
Affiliation(s)
- Mingxia Zhai
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weichen Song
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhe Liu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenxiang Cai
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guan Ning Lin
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
10
|
Li P, Tong T, Shao X, Han Y, Zhang M, Li Y, Lv X, Li H, Li Z. The synergism of Lactobacillaceae, inulin, polyglucose, and aerobic exercise ameliorates hyperglycemia by modulating the gut microbiota community and the metabolic profiles in db/db mice. Food Funct 2024; 15:4832-4851. [PMID: 38623620 DOI: 10.1039/d3fo04642g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
This study aimed to assess the impact of Lactobacillaceae (L or H represents a low or high dose), inulin (I), and polydextrose (P) combined with aerobic exercise (A) on the composition of the gut microbiota and metabolic profiles in db/db mice. After a 12-week intervention, LIP, LIPA, and HIPA groups exhibited significant improvements in hyperglycemia, glucose tolerance, insulin resistance, inflammatory response, and short-chain fatty acid (SCFA) and blood lipid levels compared to type 2 diabetes mice (MC). After treatment, the gut microbiota composition shifted favorably in the treatment groups which significantly increased the abundance of beneficial bacteria, such as Bacteroides, Blautia, Akkermansia, and Faecalibaculum, and significantly decreased the abundance of Proteus. Metabolomics analysis showed that compared to the MC group, the contents of 5-hydroxyindoleacetic acid, 3-hydroxysebacic acid, adenosine monophosphate (AMP), xanthine and hypoxanthine were significantly decreased, while 3-ketosphinganine, sphinganine, and sphingosine were significantly increased in the LIP and LIPA groups, respectively. Additionally, LIP and LIPA not only improved sphingolipid metabolism and purine metabolism pathways but also activated AMP-activated protein kinase to promote β-oxidation by increasing the levels of SCFAs. Faecalibaculum, Blautia, Bacteroides, and Akkermansia exhibited positive correlations with sphingosine, 3-ketosphinganine, and sphinganine, and exhibited negative correlations with hypoxanthine, xanthine and AMP. Faecalibaculum, Blautia, Bacteroides, and Akkermansia may have the potential to improve sphingolipid metabolism and purine metabolism pathways. These findings suggest that the synergism of Lactobacillaceae, inulin, polydextrose, and aerobic exercise provides a promising strategy for the prevention and management of type 2 diabetes.
Collapse
Affiliation(s)
- Peifan Li
- College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Tong Tong
- College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Xinyu Shao
- College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Yan Han
- College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Sino Canada Health Engineering Research Institute, Hefei, China
| | - Yongli Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xue Lv
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Hao Li
- Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450003, China.
| | - Zuming Li
- College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| |
Collapse
|
11
|
Miyamoto Y, Kikuta J, Matsui T, Hasegawa T, Fujii K, Okuzaki D, Liu YC, Yoshioka T, Seno S, Motooka D, Uchida Y, Yamashita E, Kobayashi S, Eguchi H, Morii E, Tryggvason K, Shichita T, Kayama H, Atarashi K, Kunisawa J, Honda K, Takeda K, Ishii M. Periportal macrophages protect against commensal-driven liver inflammation. Nature 2024; 629:901-909. [PMID: 38658756 DOI: 10.1038/s41586-024-07372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.
Collapse
Affiliation(s)
- Yu Miyamoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Takahiro Matsui
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Kentaro Fujii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yu-Chen Liu
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takuya Yoshioka
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Daisuke Motooka
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yutaka Uchida
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Erika Yamashita
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Karl Tryggvason
- Cardiovascular and Metabolic Disorders Program, Duke-NUS, Duke-NUS Medical School, Singapore, Singapore
| | - Takashi Shichita
- Laboratory for Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hisako Kayama
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Atarashi
- Department of Microbiology and Immunology, School of Medicine, Keio University, Tokyo, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, School of Medicine, Keio University, Tokyo, Japan
| | - Kiyoshi Takeda
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Life-omics Research Division, Institute for Open and Transdisciplinary Research Initiative, Osaka University, Osaka, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| |
Collapse
|
12
|
Yan W, Zhuang Z, Gao Y, Wang Y, He D. A Mendelian randomization investigation of the causal association between the gut microbiota and sleep disorders. Front Microbiol 2024; 15:1372827. [PMID: 38585691 PMCID: PMC10995228 DOI: 10.3389/fmicb.2024.1372827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
Background Increasing numbers of people are suffering from sleep disorders. The gut microbiota of these individuals differs significantly. However, no reports are available on the causal associations between specific gut microbiota and sleep disorders. Methods Data on gut genera were obtained from the MiBioGen consortium. Twenty-four cohorts with 18,340 individuals of European origin were included. Sleep disorder data, which included 216,454 European individuals, were retrieved from the FinnGen Biobank. Subsequently, two-sample Mendelian randomization was performed to analyze associations between sleep disorders and specific components of the gut microbiota. Results Inverse variance weighting (IVW) revealed a negative correlation between Coprobacter and sleep disorders (OR = 0.797, 95% CI = 0.66-0.96, and p = 0.016), a positive correlation between Lachnospiraceae and sleep disorders (OR = 1.429, 95% CI = 1.03-1.98, and p = 0.032), a negative association between Oscillospira and sleep disorders (OR = 0.745, 95% CI = 0.56-0.98, and p = 0.038), and a negative association between Peptococcus and sleep disorders (OR = 0.858, 95% CI = 0.74-0.99, p = 0.039). Conclusion A significant causal relationship was found between four specific gut microbiota and sleep disorders. One family, Lachnospiraceae, was observed to increase the risk of sleep disorders, while three genera, namely, Coprobacter, Oscillospira, and Peptococcus, could reduce the risk of sleep disorders. However, further investigations are needed to confirm the specific mechanisms by which the gut microbiota affects sleep.
Collapse
Affiliation(s)
- Wei Yan
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhenzhen Zhuang
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yuhao Gao
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yuntao Wang
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
| | - Daikun He
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai, China
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Szentirmai E, Buckley K, Massie AR, Kapas L. Lipopolysaccharide-Mediated Effects of the Microbiota on Sleep and Body Temperature. RESEARCH SQUARE 2024:rs.3.rs-3995260. [PMID: 38496422 PMCID: PMC10942547 DOI: 10.21203/rs.3.rs-3995260/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Background Recent research suggests that microbial molecules translocated from the intestinal lumen into the host's internal environment may play a role in various physiological functions, including sleep. Previously, we identified that butyrate, a short-chain fatty acid, produced by intestinal bacteria, and lipoteichoic acid, a cell wall component of gram-positive bacteria induce sleep when their naturally occurring translocation is mimicked by direct delivery into the portal vein. Building upon these findings, we aimed to explore the sleep signaling potential of intraportally administered lipopolysaccharide, a primary component of gram-negative bacterial cell walls, in rats. Results Low dose of lipopolysaccharide (1 μg/kg) increased sleep duration and prolonged fever, without affecting systemic lipopolysaccharide levels. Interestingly, administering LPS systemically outside the portal region at a dose 20 times higher did not affect sleep, indicating a localized sensitivity within the hepatoportal region, encompassing the portal vein and liver, for the sleep and febrile effects of lipopolysaccharide. Furthermore, both the sleep- and fever-inducing effects of LPS were inhibited by indomethacin, a prostaglandin synthesis inhibitor, and replicated by intraportal administration of prostaglandin E2 or arachidonic acid, suggesting the involvement of the prostaglandin system in mediating these actions. Conclusions These findings underscore the dynamic influence of lipopolysaccharide in the hepatoportal region on sleep and fever mechanisms, contributing to a complex microbial molecular assembly that orchestrates communication between the intestinal microbiota and brain. Lipopolysaccharide is a physiological component of plasma in both the portal and extra-portal circulation, with its levels rising in response to everyday challenges like high-fat meals, moderate alcohol intake, sleep loss and psychological stress. The increased translocation of lipopolysaccharide under such conditions may account for their physiological impact in daily life, highlighting the intricate interplay between microbial molecules and host physiology.
Collapse
|
14
|
Mio K, Iida-Tanaka N, Togo-Ohno M, Tadenuma N, Yamanaka C, Aoe S. Barley consumption under a high-fat diet suppresses lipogenic genes through altered intestinal bile acid composition. J Nutr Biochem 2024; 125:109547. [PMID: 38081474 DOI: 10.1016/j.jnutbio.2023.109547] [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: 04/20/2023] [Revised: 10/31/2023] [Accepted: 12/06/2023] [Indexed: 12/31/2023]
Abstract
We evaluated whether barley flour consumption in a high-fat environment affects lipid metabolism through signals mediated by bile acids. Four-week-old mice were fed a high-fat diet supplemented with cellulose (HC) or β-glucan-rich barley flour (HB) for 12 weeks. Bile acid composition in the intestinal tract and feces was measured by GC/MS. Gene expression levels involved in bile acid metabolism in the liver and intestinal tract were determined by RT-PCR. Similar parameters were measured in mice treated with antibiotics (antibiotics-cellulose [AC] and antibiotics-barley [AB]) to reduce the activity of intestinal bacteria. The Results showed that the HB group had lower liver blood cholesterol and triglyceride levels than the HC group. The HB group showed a significant decrease in primary bile acids in the gastrointestinal tract compared to the HC group. On the other hand, the concentration of secondary bile acids relatively increased in the cecum and feces. In the liver, Fxr activation suppressed gene expression levels in synthesizing bile acids and lipids. Furthermore, in the gastrointestinal tract, Tgr5 was activated by increased secondary bile acids. Correspondingly, AMP levels were increased in the HB group compared to the HC group, AMPK was phosphorylated in the liver, and gene expression involved in lipid synthesis was downregulated. A comparison of the AC and AB groups treated with antibiotics did not confirm these effects of barley intake. In summary, our results suggest that the prevention of lipid accumulation by barley consumption involves signaling through changes in bile acid composition in the intestinal tract.
Collapse
Affiliation(s)
- Kento Mio
- Graduate School of Studies in Human Culture, Otsuma Women's University, Tokyo, Japan; Research and Development Department, Hakubaku Co., Ltd., Yamanashi, Japan
| | - Naoko Iida-Tanaka
- Graduate School of Studies in Human Culture, Otsuma Women's University, Tokyo, Japan; The Institute of Human Culture Studies, Otsuma Women's University, Tokyo, Japan
| | - Marina Togo-Ohno
- Research and Development Department, Hakubaku Co., Ltd., Yamanashi, Japan
| | - Natsuki Tadenuma
- Graduate School of Studies in Human Culture, Otsuma Women's University, Tokyo, Japan
| | - Chiemi Yamanaka
- The Institute of Human Culture Studies, Otsuma Women's University, Tokyo, Japan
| | - Seiichiro Aoe
- Graduate School of Studies in Human Culture, Otsuma Women's University, Tokyo, Japan; The Institute of Human Culture Studies, Otsuma Women's University, Tokyo, Japan.
| |
Collapse
|
15
|
Han Y, Zeng X, Hua L, Quan X, Chen Y, Zhou M, Chuang Y, Li Y, Wang S, Shen X, Wei L, Yuan Z, Zhao Y. The fusion of multi-omics profile and multimodal EEG data contributes to the personalized diagnostic strategy for neurocognitive disorders. MICROBIOME 2024; 12:12. [PMID: 38243335 PMCID: PMC10797890 DOI: 10.1186/s40168-023-01717-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/07/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND The increasing prevalence of neurocognitive disorders (NCDs) in the aging population worldwide has become a significant concern due to subjectivity of evaluations and the lack of precise diagnostic methods and specific indicators. Developing personalized diagnostic strategies for NCDs has therefore become a priority. RESULTS Multimodal electroencephalography (EEG) data of a matched cohort of normal aging (NA) and NCDs seniors were recorded, and their faecal samples and urine exosomes were collected to identify multi-omics signatures and metabolic pathways in NCDs by integrating metagenomics, proteomics, and metabolomics analysis. Additionally, experimental verification of multi-omics signatures was carried out in aged mice using faecal microbiota transplantation (FMT). We found that NCDs seniors had low EEG power spectral density and identified specific microbiota, including Ruminococcus gnavus, Enterocloster bolteae, Lachnoclostridium sp. YL 32, and metabolites, including L-tryptophan, L-glutamic acid, gamma-aminobutyric acid (GABA), and fatty acid esters of hydroxy fatty acids (FAHFAs), as well as disturbed biosynthesis of aromatic amino acids and TCA cycle dysfunction, validated in aged mice. Finally, we employed a support vector machine (SVM) algorithm to construct a machine learning model to classify NA and NCDs groups based on the fusion of EEG data and multi-omics profiles and the model demonstrated 92.69% accuracy in classifying NA and NCDs groups. CONCLUSIONS Our study highlights the potential of multi-omics profiling and EEG data fusion in personalized diagnosis of NCDs, with the potential to improve diagnostic precision and provide insights into the underlying mechanisms of NCDs. Video Abstract.
Collapse
Affiliation(s)
- Yan Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xinglin Zeng
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Lin Hua
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xingping Quan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Ying Chen
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Manfei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | | | - Yang Li
- Department of Gastrointestinal Surgery, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lai Wei
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Zhen Yuan
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China.
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078, Macau SAR, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China.
| |
Collapse
|
16
|
Tayama J, Hamaguchi T, Koizumi K, Yamamura R, Okubo R, Kawahara JI, Inoue K, Takeoka A, Fukudo S. Efficacy of an eHealth self-management program in reducing irritable bowel syndrome symptom severity: a randomized controlled trial. Sci Rep 2024; 14:4. [PMID: 38172498 PMCID: PMC10764726 DOI: 10.1038/s41598-023-50293-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
This study aimed to verify whether an eHealth-based self-management program can reduce irritable bowel syndrome (IBS) symptom severity. An open-label simple randomized controlled trial was conducted that compared an intervention group (n = 21) participating in an eHealth self-management program, which involved studying IBS-related information from an established self-help guide followed by in-built quizzes, with a treatment-as-usual group (n = 19) that, except for pharmacotherapy, had no treatment restrictions. Participants were female Japanese university students. The eHealth group received unlimited access to the self-management program for 8 weeks on computers and mobile devices. The primary outcome, participants' severity of IBS symptoms assessed using the IBS-severity index (IBS-SI), and the secondary outcomes of participants' quality of life, gut bacteria, and electroencephalography alpha and beta power percentages were measured at baseline and 8 weeks. A significant difference was found in the net change in IBS-SI scores between the eHealth and treatment-as-usual groups, and the former had significantly lower IBS-SI scores following the 8-week intervention than at baseline. Moreover, there was a significant difference in the net change in phylum Cyanobacteria between the eHealth and treatment-as-usual groups. Thus, the eHealth-based self-management program successfully reduced the severity of IBS symptoms.
Collapse
Affiliation(s)
- Jun Tayama
- Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama, 359-1192, Japan.
| | - Toyohiro Hamaguchi
- Department of Occupational Therapy, School of Health and Social Services, Saitama Prefectural University, 820, Sannomiya, Koshigaya, Saitama, 343-8540, Japan
| | - Kohei Koizumi
- Department of Occupational Therapy, School of Health and Social Services, Saitama Prefectural University, 820, Sannomiya, Koshigaya, Saitama, 343-8540, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-0815, Japan
| | - Ryo Okubo
- Department of Psychiatry and Neurology, National Hospital Organization Obihiro Hospital, 16, Kita 2, Nishi 18, Obihiro, Hokkaido, 080-0048, Japan
| | - Jun-Ichiro Kawahara
- Department of Psychology, Hokkaido University, Kita 10, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-0815, Japan
| | - Kenji Inoue
- Center for Student Success Research and Practice, Osaka University, 1-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Takeoka
- Health Center, Nagasaki University, 1-14, Bunkyo, Nagasaki, Nagasaki, 852-8521, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
17
|
Gu L, Ni Y, Wang B, Kong L, Yu S, Tang Y, Zhu P, Shao S, Tao F, Liu K. Antibiotic exposure associated with nighttime sleep duration and daytime sleepiness in newlyweds. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:6350-6371. [PMID: 38148462 DOI: 10.1007/s11356-023-31475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
Few studies have explored the relationship between antibiotic exposure and sleep in newlyweds. We applied the actor-partner interdependence moderation model to estimate the relationships of antibiotic exposure with nighttime sleep duration (weekday, weekend, and average sleep durations) and daytime sleepiness in newlyweds. We found that 99.0% of the 2698 enrolled individuals were exposed to at least one antibiotic. Among the newlyweds, exposure to florfenicol (β, - 0.077; 95% confidence interval [CI], - 0.143, - 0.011), exposure to chloramphenicols (- 0.086 [- 0.160, - 0.011]), and exposure to veterinary antibiotics (VAs) (- 0.106 [- 0.201, - 0.010]) were negatively associated with weekday sleep duration. Florfenicol, chloramphenicols, and VAs were also inversely related to average sleep duration in the newlyweds. Ciprofloxacin and cyadox exposure was significantly associated with an increase of 0.264 (0.030, 0.497) and (0.375 [0.088, 0.663]) Epworth Sleepiness Scale (ESS) scores in the newlyweds, respectively. Gender moderated the actor-partner effects of erythromycin and tetracyclines on the newlyweds' weekday sleep duration and ESS scores. Overall, exposure to florfenicol, chloramphenicols, and VAs shortened weekday and average sleep durations of newlyweds. Exposure to ciprofloxacin and cyadox promoted daytime sleepiness. Gender moderated the actor-partner effects of specific antibiotics on the weekday sleep duration and ESS scores of the newlyweds.
Collapse
Affiliation(s)
- Lvfen Gu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yachao Ni
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Baolin Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li Kong
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shuixin Yu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ying Tang
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peng Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shanshan Shao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
18
|
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
|
19
|
Jezkova J, Sonka K, Kreisinger J, Prochazkova P, Tlaskalova-Hogenova H, Nevsimalova S, Buskova J, Merkova R, Dvorakova T, Prihodova I, Dostalova S, Roubalova R. Guardians of Rest? Investigating the gut microbiota in central hypersomnolence disorders. Sleep Med 2024; 113:95-102. [PMID: 37995475 DOI: 10.1016/j.sleep.2023.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
In recent years, there has been an increased interest in elucidating the influence of the gut microbiota on sleep physiology. The gut microbiota affects the central nervous system by modulating neuronal pathways through the neuroendocrine and immune system, the hypothalamus-pituitary-adrenal axis, and various metabolic pathways. The gut microbiota can also influence circadian rhythms. In this study, we observed the gut microbiota composition of patients suffering from narcolepsy type 1, narcolepsy type 2, and idiopathic hypersomnia. We did not observe any changes in the alpha diversity of the gut microbiota among patient groups and healthy controls. We observed changes in beta diversity in accordance with Jaccard dissimilarities between the control group and groups of patients suffering from narcolepsy type 1 and idiopathic hypersomnia. Our results indicate that both these patient groups differ from controls relative to the presence of rare bacterial taxa. However, after adjustment for various confounding factors such as BMI, age, and gender, there were no statistical differences among the groups. This indicates that the divergence in beta diversity in the narcolepsy type 1 and idiopathic hypersomnia groups did not arise due to sleep disturbances. This study implies that using metabolomics and proteomics approaches to study the role of microbiota in sleep disorders might prove beneficial.
Collapse
Affiliation(s)
- Janet Jezkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jakub Kreisinger
- Faculty of Science, Department of Zoology, Charles University, Prague, Czech Republic
| | - Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Sona Nevsimalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jitka Buskova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radana Merkova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Dvorakova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Iva Prihodova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Simona Dostalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
20
|
Tanihiro R, Yuki M, Sasai M, Haseda A, Kagami-Katsuyama H, Hirota T, Honma N, Nishihira J. Effects of Prebiotic Yeast Mannan on Gut Health and Sleep Quality in Healthy Adults: A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2023; 16:141. [PMID: 38201970 PMCID: PMC10780920 DOI: 10.3390/nu16010141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Human gut health is closely related to sleep. We aimed to evaluate the efficacy of yeast mannan (YM) in improving bowel habits and sleep quality, along with metabolomics in fecal samples. A total of 40 healthy adults (age range, 22-64 years) with discomfort in defecation were enrolled and randomly allocated to receive either YM (n = 20; 1.1 g/day) or placebo (n = 20) for four weeks. Participants recorded their defecation habits throughout the test periods. Sleep electroencephalogram (EEG) recording using an EEG device and fecal sampling were performed pre- and post-treatment. The YM group significantly increased defecation frequency and stool volumes compared to the placebo group. After 4 weeks of treatment, the non-REM sleep stage 3 (N3) duration in the YM group was significantly higher than that in the placebo group. YM ingestion significantly lengthened total time in bed (TIB) and significantly shortened N3 latency compared to placebo intake during the trial. The metabolomics analysis found a total of 20 metabolite differences between the YM and placebo groups. As a result of stepwise linear regression, changes in fecal propionate and gamma-aminobutyric acid (GABA) levels were identified as the primary factors explaining changes in TIB and N3 latency, respectively. Our findings suggest that the prebiotic YM could be beneficial to gut health and sleep quality.
Collapse
Affiliation(s)
- Reiko Tanihiro
- Core Technology Laboratories, Asahi Quality and Innovations, Ltd., Moriya 302-0106, Japan; (M.Y.); (M.S.); (T.H.)
| | - Masahiro Yuki
- Core Technology Laboratories, Asahi Quality and Innovations, Ltd., Moriya 302-0106, Japan; (M.Y.); (M.S.); (T.H.)
| | - Masaki Sasai
- Core Technology Laboratories, Asahi Quality and Innovations, Ltd., Moriya 302-0106, Japan; (M.Y.); (M.S.); (T.H.)
| | - Akane Haseda
- Department of Medical Management and Informatics, Hokkaido Information University, Ebetsu 069-8585, Japan (J.N.)
| | - Hiroyo Kagami-Katsuyama
- Department of Medical Management and Informatics, Hokkaido Information University, Ebetsu 069-8585, Japan (J.N.)
| | - Tatsuhiko Hirota
- Core Technology Laboratories, Asahi Quality and Innovations, Ltd., Moriya 302-0106, Japan; (M.Y.); (M.S.); (T.H.)
| | - Naoyuki Honma
- Department of Medical Management and Informatics, Hokkaido Information University, Ebetsu 069-8585, Japan (J.N.)
| | - Jun Nishihira
- Department of Medical Management and Informatics, Hokkaido Information University, Ebetsu 069-8585, Japan (J.N.)
| |
Collapse
|
21
|
Ehichioya DE, Taufique SKT, Magaña I, Farah S, Obata Y, Yamazaki S. Gut microbiota depletion minimally affects the daily voluntary wheel running activity and food anticipatory activity in female and male C57BL/6J mice. Front Physiol 2023; 14:1299474. [PMID: 38107475 PMCID: PMC10722266 DOI: 10.3389/fphys.2023.1299474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Emerging evidence has highlighted that the gut microbiota plays a critical role in the regulation of various aspects of mammalian physiology and behavior, including circadian rhythms. Circadian rhythms are fundamental behavioral and physiological processes that are governed by circadian pacemakers in the brain. Since mice are nocturnal, voluntary wheel running activity mostly occurs at night. This nocturnal wheel-running activity is driven by the primary circadian pacemaker located in the suprachiasmatic nucleus (SCN). Food anticipatory activity (FAA) is the increased bout of locomotor activity that precedes the scheduled short duration of a daily meal. FAA is controlled by the food-entrainable oscillator (FEO) located outside of the SCN. Several studies have shown that germ-free mice and mice with gut microbiota depletion altered those circadian behavioral rhythms. Therefore, this study was designed to test if the gut microbiota is involved in voluntary wheel running activity and FAA expression. To deplete gut microbiota, C57BL/6J wildtype mice were administered an antibiotic cocktail via their drinking water throughout the experiment. The effect of antibiotic cocktail treatment on wheel running activity rhythm in both female and male mice was not detectable with the sample size in our current study. Then mice were exposed to timed restricted feeding during the day. Both female and male mice treated with antibiotics exhibited normal FAA which was comparable with the FAA observed in the control group. Those results suggest that gut microbiota depletion has minimum effect on both circadian behavioral rhythms controlled by the SCN and FEO respectively. Our result contradicts recently published studies that reported significantly higher FAA levels in germ-free mice compared to their control counterparts and gut microbiota depletion significantly reduced voluntary activity by 50%.
Collapse
Affiliation(s)
- David E. Ehichioya
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
| | | | - Isabel Magaña
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Sofia Farah
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
| | - Yuuki Obata
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Shin Yamazaki
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
22
|
Singh S, Mahajan M, Kumar D, Singh K, Chowdhary M, Amit. An inclusive study of recent advancements in Alzheimer's disease: A comprehensive review. Neuropeptides 2023; 102:102369. [PMID: 37611472 DOI: 10.1016/j.npep.2023.102369] [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: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) has remained elusive in revealing its pathophysiology and mechanism of development. In this review paper, we attempt to highlight several theories that abound about the exact pathway of AD development. The number of cases worldwide has prompted a constant flow of research to detect high-risk patients, slow the progression of the disease and discover improved methods of treatment that may prove effective. We shall focus on the two main classes of drugs that are currently in use; and emerging ones with novel mechanisms that are under development. As of late there has also been increased attention towards factors that were previously thought to be unrelated to AD, such as the gut microbiome, lifestyle habits, and diet. Studies have now shown that all these factors make an impact on AD progression, thus bringing to our attention more areas that could hold the key to combating this disease. This paper covers all the aforementioned factors concisely. We also briefly explore the relationship between mental health and AD, both before and after the diagnosis of the disease.
Collapse
Affiliation(s)
- Sukanya Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mitali Mahajan
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Dhawal Kumar
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Kunika Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mehvish Chowdhary
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Amit
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India.
| |
Collapse
|
23
|
Fang H, Yao T, Li W, Pan N, Xu H, Zhao Q, Su Y, Xiong K, Wang J. Efficacy and safety of fecal microbiota transplantation for chronic insomnia in adults: a real world study. Front Microbiol 2023; 14:1299816. [PMID: 38088972 PMCID: PMC10712199 DOI: 10.3389/fmicb.2023.1299816] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/08/2023] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE To assess the efficacy and safety of fecal microbiota transplantation (FMT) for adult chronic insomnia. METHODS Patients treated with FMT for chronic diseases were divided into chronic insomnia and non-insomnia group. The primary endpoint was the efficacy of FMT for insomnia 4 weeks after treatment, the secondary endpoints included the impacts of FMT on anxiety, depression, health-related quality of life, gut microbiota, and adverse events associated with FMT. Insomnia Severity Index (ISI) and Pittsburgh Sleep Quality Index (PSQI) were utilized to assess the efficacy of FMT on insomnia, self-rating anxiety/depression scale [Zung Self-Rating Anxiety Scale (SAS), Zung Self-Rating Depression Scale (SDS)] was employed to evaluate anxiety and depression. Quality of life was evaluated by SF-36. 16S rRNA sequencing was employed to analyze the gut microbiota and correlation analysis was performed. RESULTS Forty patients met the inclusion criteria and seven were excluded. 33 patients were enrolled and stratified into chronic insomnia group (N = 17) and non-insomnia group (N = 16). Compared to baseline, FMT significantly ameliorated the ISI (17.31 ± 5.12 vs. 5.38 ± 5.99), PSQI (14.56 ± 2.13 vs. 6.63 ± 4.67), SAS (54.25 ± 8.90 vs. 43.68 ± 10.64) and SDS (57.43 ± 10.96 vs. 50.68 ± 15.27) score and quality of life of chronic insomnia patients. 76.47% (13/17) of insomnia patients achieved the primary endpoints. In chronic insomnia patients, the relative abundance of Eggerthella marked enhanced at baseline, while the relative abundance of Lactobacillus, Bifidobacterium, Turicibacter, Anaerostipes, and Eisenbergiella significantly increased after FMT treatment, the latter positive correlated with the efficacy of FMT. Encouragingly, FMT also improved the sleep quality of non-insomnia patients. CONCLUSION Eggerthella may potentially serve as a distinctive genus associated with chronic insomnia. FMT maybe a novel treatment option for adults with chronic insomnia and provide an alternative to traditional treatments for insomnia. The effects were positive correlated with the augmentation of probiotics, such as Bifidobacterium, Lactobacillus, Turicibacter, and Fusobacterium.
Collapse
Affiliation(s)
- Haiming Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Diagnosis and Treatment, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tingting Yao
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Diagnosis and Treatment, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wanli Li
- Department of Pharmacology, School of Basic Medical Sciences of Anhui Medical University, Hefei, Anhui, China
| | - Na Pan
- Department of Pharmacology, School of Basic Medical Sciences of Anhui Medical University, Hefei, Anhui, China
| | - Hang Xu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
| | - Qian Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Diagnosis and Treatment, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuan Su
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Diagnosis and Treatment, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kangwei Xiong
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Diagnosis and Treatment, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiajia Wang
- Department of Pharmacology, School of Basic Medical Sciences of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
24
|
Arnoriaga-Rodríguez M, Leal Y, Mayneris-Perxachs J, Pérez-Brocal V, Moya A, Ricart W, Fernández-Balsells M, Fernández-Real JM. Gut Microbiota Composition and Functionality Are Associated With REM Sleep Duration and Continuous Glucose Levels. J Clin Endocrinol Metab 2023; 108:2931-2939. [PMID: 37159524 DOI: 10.1210/clinem/dgad258] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/18/2023] [Accepted: 05/05/2023] [Indexed: 05/11/2023]
Abstract
CONTEXT Sleep disruption is associated with worse glucose metabolic control and altered gut microbiota in animal models. OBJECTIVE We aimed to evaluate the possible links among rapid eye movement (REM) sleep duration, continuous glucose levels, and gut microbiota composition. METHODS This observational, prospective, real-life, cross-sectional case-control study included 118 (60 with obesity), middle-aged (39.1-54.8 years) healthy volunteers recruited at a tertiary hospital. Glucose variability and REM sleep duration were assessed by 10-day continuous glucose monitoring (CGM) (Dexcom G6) and wrist actigraphy (Fitbit Charge 3), respectively. The coefficient of variation (CV), interquartile range (IQR), and SD of glucose variability was assessed and the percentage of time in range (% TIR), at 126-139 mg/dL (TIR2), and 140-199 mg/dL (TIR3) were calculated. Shotgun metagenomics sequencing was applied to study gut microbiota taxonomy and functionality. RESULTS Increased glycemic variability (SD, CV, and IQR) was observed among subjects with obesity in parallel to increased % TIR2 and % TIR3. REM sleep duration was independently associated with % TIR3 (β = -.339; P < .001) and glucose variability (SD, β = -.350; P < .001). Microbial taxa from the Christensenellaceae family (Firmicutes phylum) were positively associated with REM sleep and negatively with CGM levels, while bacteria from Enterobacteriacea family and bacterial functions involved in iron metabolism showed opposite associations. CONCLUSION Decreased REM sleep duration was independently associated with a worse glucose profile. The associations of species from Christensenellaceae and Enterobacteriaceae families with REM sleep duration and continuous glucose values suggest an integrated picture of metabolic health.
Collapse
Affiliation(s)
- María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Yenny Leal
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Vicente Pérez-Brocal
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Andrés Moya
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), 46020 Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC), 46980 Valencia, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Mercè Fernández-Balsells
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| |
Collapse
|
25
|
Estaki M, Langsetmo L, Shardell M, Mischel A, Jiang L, Zhong Y, Kaufmann C, Knight R, Stone K, Kado D. Association of Subjective and Objective Measures of Sleep With Gut Microbiota Composition and Diversity in Older Men: The Osteoporotic Fractures in Men Study. J Gerontol A Biol Sci Med Sci 2023; 78:1925-1932. [PMID: 36655399 PMCID: PMC10562887 DOI: 10.1093/gerona/glad011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Growing evidence suggests bidirectional links between gut microbiota and sleep quality as shared contributors to health. Little is known about the relationship between microbiota and sleep among older persons. METHODS We used 16S rRNA sequencing to characterize stool microbiota among men (n = 606, mean [standard deviation] age = 83.9 [3.8]) enrolled in the Osteoporotic Fractures in Men (MrOS) study from 2014 to 2016. Sleep was assessed concurrently by a questionnaire (Pittsburgh Sleep Quality index [PSQI]), and activity monitor to examine timing (acrophase) and regularity of patterns (F-statistic). Alpha diversity was measured using Faith's phylogenetic diversity (PD). Beta diversity was calculated with robust Aitchison distance with matrix completion (RPCA) and phylogenetic-RPCA (PRPCA). Their association with sleep variables was tested with partial distance-based redundancy analysis (dbRDA). Predictive-ratio biomarkers associated with sleep measurements were identified with CoDaCoRe. RESULTS In unadjusted analyses, men with poor sleep (PSQI >5) tended to have lower alpha diversity compared to men with normal sleep (Faith's PD, beta = -0.15; 95% confidence interval [CI]: -0.30 to 0.01, p = .06). Sleep regularity was significantly associated with RPCA and PRPCA, even after adjusting for site, batch, age, ethnicity, body mass index, diabetes, antidepressant and sleep medication use, and health behaviors (RPCA/PRPCA dbRDA; p = .033/.002). In taxonomic analysis, ratios of 7:6 bacteria for better regularity (p = .0004) and 4:7 for worse self-reported sleep (p = .005) were differentially abundant: some butyrate-producing bacteria were associated with better sleep characteristics. CONCLUSIONS Subjective and objective indicators of sleep quality suggest that older men with better sleep patterns are more likely to harbor butyrate-producing bacteria associated with better health.
Collapse
Affiliation(s)
- Mehrbod Estaki
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Lisa Langsetmo
- Center for Care Delivery and Outcomes Research, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Michelle Shardell
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna Mischel
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lingjing Jiang
- Janssen Research and Development Los Angeles, Los Angeles, California, USA
| | - Yuan Zhong
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Christopher Kaufmann
- Department of Aging and Geriatric Research, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Rob Knight
- Department of Pediatrics, UC San Diego, La Jolla, California, USA
- UC San Diego Center for Microbiome Innovation, La Jolla, California, USA
- Department of Computer Science and Engineering, UC San Diego, La Jolla, California, USA
- Department of Bioengineering, UC San Diego, La Jolla, California, USA
| | - Katie Stone
- Department of Research, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Deborah Kado
- Geriatrics Section, Stanford University School of Medicine, Palo Alto, California, USA
- Veterans Health Administration, Geriatrics Research Education and Clinical Center, Palo Alto, California, USA
| |
Collapse
|
26
|
Mühlematter C, Nielsen DS, Castro-Mejía JL, Brown SA, Rasch B, Wright KP, Walser JC, Schoch SF, Kurth S. Not simply a matter of parents-Infants' sleep-wake patterns are associated with their regularity of eating. PLoS One 2023; 18:e0291441. [PMID: 37796923 PMCID: PMC10553286 DOI: 10.1371/journal.pone.0291441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/28/2023] [Indexed: 10/07/2023] Open
Abstract
In adults there are indications that regular eating patterns are related to better sleep quality. During early development, sleep and eating habits experience major maturational transitions. Further, the bacterial landscape of the gut microbiota undergoes a rapid increase in complexity. Yet little is known about the association between sleep, eating patterns and the gut microbiota. We first hypothesized that higher eating regularity is associated with more mature sleep patterns, and second, that this association is mediated by the maturational status of the gut microbiota. To test this hypothesis, we performed a longitudinal study in 162 infants to assess actigraphy, diaries of sleep and eating times, and stool microbiota composition at ages 3, 6 and 12 months. To comprehensively capture infants' habitual sleep-wake patterns, 5 sleep composites that characterize infants' sleep habits across multiple days in their home environment were computed. To assess timing of eating habits, we developed an Eating Regularity Index (ERI). Gut microbial composition was assessed by 16S rRNA gene amplicon sequencing, and its maturation was assessed based on alpha diversity, bacterial maturation index, and enterotype. First, our results demonstrate that increased eating regularity (higher ERI) in infants is associated with less time spent awake during the night (sleep fragmentation) and more regular sleep patterns. Second, the associations of ERI with sleep evolve with age. Third, the link between infant sleep and ERI remains significant when controlling for parents' subjectively rated importance of structuring their infant's eating and sleeping times. Finally, the gut microbial maturational markers did not account for the link between infant's sleep patterns and ERI. Thus, infants who eat more regularly have more mature sleep patterns, which is independent of the maturational status of their gut microbiota. Interventions targeting infant eating rhythm thus constitute a simple, ready-to-use anchor to improve sleep quality.
Collapse
Affiliation(s)
| | - Dennis S. Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Josue L. Castro-Mejía
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Steven A. Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Björn Rasch
- Department of Psychology, University of Fribourg, Fribourg, Switzerland
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | | | - Sarah F. Schoch
- Donders Institute for Brain, Radboud University Medical Center, Nijmegen, Netherlands
| | - Salome Kurth
- Department of Psychology, University of Fribourg, Fribourg, Switzerland
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Post Z, Manfready RA, Keshavarzian A. Overview of the Gut-Brain Axis: From Gut to Brain and Back Again. Semin Neurol 2023; 43:506-517. [PMID: 37562457 DOI: 10.1055/s-0043-1771464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The gut-brain axis refers to a bidirectional communication pathway linking the gastrointestinal system to the central nervous system. The hardware of this multifaceted pathway takes many forms, at once structural (neurons, microglia, intestinal epithelial cell barrier), chemical (neurotransmitters, enteroendocrine hormones, bacterial metabolites), and cellular (immune signaling, inflammatory pathways). The gut-brain axis is exquisitely influenced by our environment, diet, and behaviors. Here, we will describe recent progress in understanding the gut-brain axis in neurological disease, using Parkinson's disease as a guide. We will see that each component of the gut-brain axis is heavily mediated by intestinal microbiota and learn how gut-brain communication can go awry in microbial dysbiosis.
Collapse
Affiliation(s)
- Zoë Post
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Richard A Manfready
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois
- Departments of Physiology and Anatomy & Cell Biology, Rush University Medical Center, Chicago, Illinois
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois
- Departments of Physiology and Anatomy & Cell Biology, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
28
|
Zhuang H, Fujikura Y, Ohkura N, Higo-Yamamoto S, Mishima T, Oishi K. A ketogenic diet containing medium-chain triglycerides reduces REM sleep duration without significant influence on mouse circadian phenotypes. Food Res Int 2023; 169:112852. [PMID: 37254426 DOI: 10.1016/j.foodres.2023.112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 06/01/2023]
Abstract
Ketogenic diets (KDs) affect the circadian rhythms of behavior and clock gene expression in experimental animals. However, these diets were designed to simulate a fasting state; thus, whether these effects are caused by diet-induced ketogenesis or persistent starvation is difficult to distinguish. The present study aimed to define the effects of a KD containing medium-chain triglycerides (MCT-KD) that increase blood ketone levels without inducing carbohydrate starvation, on circadian rhythms and sleep regulation. Mice were fed with a normal diet (CTRL) or MCT-KD for 2 weeks. Blood β-hydroxybutyrate levels were significantly increased up to 2 mM by the MCT-KD, whereas body weight gain and blood glucose levels were identical between the groups, suggesting that ketosis accumulated without carbohydrate starvation in the MCT-KD mice. Circadian rhythms of wheel-running activity and core body temperature were almost identical, although wheel-running was slightly reduced in the MCT-KD mice. The circadian expression of the core clock genes, Per1, Per2, Bmal1, and Dbp in the hypothalamus, heart, liver, epididymal adipose tissues, and skeletal muscle were almost identical between the CTRL and MCT-KD mice, whereas the amplitude of hepatic Per2 and adipose Per1 expression was increased in MCT-KD mice. The MCT-KD reduced the duration of rapid-eye-movement (REM) sleep without affecting the duration of non-REM sleep and the duration of wakefulness. These findings suggested that the impact of ketone bodies on circadian systems are limited, although they might reduce locomotor activity and REM sleep duration.
Collapse
Affiliation(s)
- Haotong Zhuang
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yuri Fujikura
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Naoki Ohkura
- Laboratory of Host Defense, School of Pharma-Sciences, Teikyo University, Itabashi, Tokyo, Japan
| | - Sayaka Higo-Yamamoto
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Taiga Mishima
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan; Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan; School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
29
|
Wang J, Zhou T, Liu F, Huang Y, Xiao Z, Qian Y, Zhou W. Influence of gut microbiota on resilience and its possible mechanisms. Int J Biol Sci 2023; 19:2588-2598. [PMID: 37215996 PMCID: PMC10197883 DOI: 10.7150/ijbs.82362] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
Excessive stress leads to disruptions of the central nervous system. Individuals' responses to stress and trauma differ from person to person. Some may develop various neuropsychiatric disorders, such as post-traumatic stress disorder, major depression, and anxiety disorders, while others may successfully adapt to the same stressful events. These two neural phenotypes are called susceptibility and resilience. Previous studies have suggested resilience/susceptibility as a complex, non-specific systemic response involving central and peripheral systems. Emerging research of mechanisms underlying resilience is mostly focussing on the physiological adaptation of specific brain circuits, neurovascular impairment of the blood-brain barrier, the role of innate and adaptive factors of the immune system, and the dysbiosis of gut microbiota. In accordance with the microbiota-gut-brain axis hypothesis, the gut microbiome directly influences the interface between the brain and the periphery to affect neuronal function. This review explored several up-to-date studies on the role of gut microbiota implicated in stressful events-related resilience/susceptibility. We mainly focus on the changes in behavior and neuroimaging characteristics, involved brain regions and circuits, the blood-brain barrier, the immune system, and epigenetic modifications, which contribute to stress-induced resilience and susceptibility. The perspective of the gut-brain axis could help to understand the mechanisms underlying resilience and the discovery of biomarkers may lead to new research directions and therapeutic interventions for stress-induced neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Ting Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yan Huang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yan Qian
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| |
Collapse
|
30
|
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: 7] [Impact Index Per Article: 3.5] [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
|
31
|
Gut Microbiota and Metabolites May Play a Crucial Role in Sea Cucumber Apostichopus japonicus Aestivation. Microorganisms 2023; 11:microorganisms11020416. [PMID: 36838381 PMCID: PMC9961660 DOI: 10.3390/microorganisms11020416] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
The constant increase in temperatures under global warming has led to a prolonged aestivation period for Apostichopus japonicus, resulting in considerable losses in production and economic benefits. However, the specific mechanism of aestivation has not been fully elucidated. In this study, we first tried to illustrate the biological mechanisms of aestivation from the perspective of the gut microbiota and metabolites. Significant differences were found in the gut microbiota of aestivating adult A. japonicus (AAJSD group) compared with nonaestivating adult A. japonicus (AAJRT group) and young A. japonicus (YAJRT and YAJSD groups) based on 16S rRNA gene high-throughput sequencing analysis. The abundances of Desulfobacterota, Myxococcota, Bdellovibrionota, and Firmicutes (4 phyla) in the AAJSD group significantly increased. Moreover, the levels of Pseudoalteromonas, Fusibacter, Labilibacter, Litorilituus, Flammeovirga, Polaribacter, Ferrimonas, PB19, and Blfdi19 genera were significantly higher in the AAJSD group than in the other three groups. Further analysis of the LDA effect size showed that species with significant variation in abundance in the AAJSD group, including the phylum Firmicutes and the genera Litorilituus, Fusibacter, and Abilibacter, might be important biomarkers for aestivating adult A. japonicus. In addition, the results of metabolomics analysis showed that there were three distinct metabolic pathways, namely biosynthesis of secondary metabolites, tryptophan metabolism, and sesquiterpenoid and triterpenoid biosynthesis in the AAJSD group compared with the other three groups. Notably, 5-hydroxytryptophan was significantly upregulated in the AAJSD group in the tryptophan metabolism pathway. Moreover, the genera Labilibacter, Litorilituus, Ferrimonas, Flammeovirga, Blfdi19, Fusibacter, Pseudoalteromonas, and PB19 with high abundance in the gut of aestivating adult A. japonicus were positively correlated with the metabolite 5-HTP. These findings suggest that there may be potential biological associations among the gut microbiota, metabolites, and aestivation in A. japonicus. This work may provide a new perspective for further understanding the aestivation mechanism of A. japonicus.
Collapse
|
32
|
Chandra S, Sisodia SS, Vassar RJ. The gut microbiome in Alzheimer's disease: what we know and what remains to be explored. Mol Neurodegener 2023; 18:9. [PMID: 36721148 PMCID: PMC9889249 DOI: 10.1186/s13024-023-00595-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/06/2023] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, results in a sustained decline in cognition. There are currently few effective disease modifying therapies for AD, but insights into the mechanisms that mediate the onset and progression of disease may lead to new, effective therapeutic strategies. Amyloid beta oligomers and plaques, tau aggregates, and neuroinflammation play a critical role in neurodegeneration and impact clinical AD progression. The upstream modulators of these pathological features have not been fully clarified, but recent evidence indicates that the gut microbiome (GMB) may have an influence on these features and therefore may influence AD progression in human patients. In this review, we summarize studies that have identified alterations in the GMB that correlate with pathophysiology in AD patients and AD mouse models. Additionally, we discuss findings with GMB manipulations in AD models and potential GMB-targeted therapeutics for AD. Lastly, we discuss diet, sleep, and exercise as potential modifiers of the relationship between the GMB and AD and conclude with future directions and recommendations for further studies of this topic.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sangram S. Sisodia
- Department of Neurobiology, University of Chicago, Chicago, IL 60637 USA
| | - Robert J. Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
33
|
Shimizu Y, Yamamura R, Yokoi Y, Ayabe T, Ukawa S, Nakamura K, Okada E, Imae A, Nakagawa T, Tamakoshi A, Nakamura K. Shorter sleep time relates to lower human defensin 5 secretion and compositional disturbance of the intestinal microbiota accompanied by decreased short-chain fatty acid production. Gut Microbes 2023; 15:2190306. [PMID: 36945116 PMCID: PMC10038026 DOI: 10.1080/19490976.2023.2190306] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Sleep is essential for our health. Short sleep is known to increase disease risks via imbalance of intestinal microbiota, dysbiosis. However, mechanisms by which short sleep induces dysbiosis remain unknown. Small intestinal Paneth cell regulates the intestinal microbiota by secreting antimicrobial peptides including α-defensin, human defensin 5 (HD5). Disruption of circadian rhythm mediating sleep-wake cycle induces Paneth cell failure. We aim to clarify effects of short sleep on HD5 secretion and the intestinal microbiota. Fecal samples and self-reported sleep time were obtained from 35 healthy middle-aged Japanese (41 to 60-year-old). Shorter sleep time was associated with lower fecal HD5 concentration (r = 0.354, p = 0.037), lower centered log ratio (CLR)-transformed abundance of short-chain fatty acid (SCFA) producers in the intestinal microbiota such as [Ruminococcus] gnavus group (r = 0.504, p = 0.002) and Butyricicoccus (r = 0.484, p = 0.003), and lower fecal SCFA concentration. Furthermore, fecal HD5 positively correlated with the abundance of these genera and SCFA concentration. These findings suggest that short sleep relates to disturbance of the intestinal microbiota via decreased HD5 secretion.
Collapse
Affiliation(s)
- Yu Shimizu
- Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Yuki Yokoi
- Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Shigekazu Ukawa
- Department of Social Welfare Science and Clinical Psychology, Osaka Metropolitan University Graduate School of Human Life and Ecology, Osaka, Japan
| | - Koshi Nakamura
- Department of Public Health and Hygiene, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Emiko Okada
- Department of Nutritional Epidemiology and Shokuiku, National Institute of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | | | | | - Akiko Tamakoshi
- Department of Public Health, Faculty of Medicine, Hokkaido University, Hokkaido, Japan
| | - Kiminori Nakamura
- Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
34
|
Diamanti T, Prete R, Battista N, Corsetti A, De Jaco A. Exposure to Antibiotics and Neurodevelopmental Disorders: Could Probiotics Modulate the Gut-Brain Axis? Antibiotics (Basel) 2022; 11:1767. [PMID: 36551423 PMCID: PMC9774196 DOI: 10.3390/antibiotics11121767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
In order to develop properly, the brain requires the intricate interconnection of genetic factors and pre-and postnatal environmental events. The gut-brain axis has recently raised considerable interest for its involvement in regulating the development and functioning of the brain. Consequently, alterations in the gut microbiota composition, due to antibiotic administration, could favor the onset of neurodevelopmental disorders. Literature data suggest that the modulation of gut microbiota is often altered in individuals affected by neurodevelopmental disorders. It has been shown in animal studies that metabolites released by an imbalanced gut-brain axis, leads to alterations in brain function and deficits in social behavior. Here, we report the potential effects of antibiotic administration, before and after birth, in relation to the risk of developing neurodevelopmental disorders. We also review the potential role of probiotics in treating gastrointestinal disorders associated with gut dysbiosis after antibiotic administration, and their possible effect in ameliorating neurodevelopmental disorder symptoms.
Collapse
Affiliation(s)
- Tamara Diamanti
- Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, 00185 Rome, Italy
| | - Roberta Prete
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Natalia Battista
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Aldo Corsetti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Antonella De Jaco
- Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
35
|
Ouyang J, Peng Y, Gong Y. New Perspectives on Sleep Regulation by Tea: Harmonizing Pathological Sleep and Energy Balance under Stress. Foods 2022; 11:3930. [PMID: 36496738 PMCID: PMC9738644 DOI: 10.3390/foods11233930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/09/2022] Open
Abstract
Sleep, a conservative evolutionary behavior of organisms to adapt to changes in the external environment, is divided into natural sleep, in a healthy state, and sickness sleep, which occurs in stressful environments or during illness. Sickness sleep plays an important role in maintaining energy homeostasis under an injury and promoting physical recovery. Tea, a popular phytochemical-rich beverage, has multiple health benefits, including lowering stress and regulating energy metabolism and natural sleep. However, the role of tea in regulating sickness sleep has received little attention. The mechanism underlying tea regulation of sickness sleep and its association with the maintenance of energy homeostasis in injured organisms remains to be elucidated. This review examines the current research on the effect of tea on sleep regulation, focusing on the function of tea in modulating energy homeostasis through sickness sleep, energy metabolism, and damage repair in model organisms. The potential mechanisms underlying tea in regulating sickness sleep are further suggested. Based on the biohomology of sleep regulation, this review provides novel insights into the role of tea in sleep regulation and a new perspective on the potential role of tea in restoring homeostasis from diseases.
Collapse
Affiliation(s)
- Jin Ouyang
- Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Yuxuan Peng
- Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
- College of Physical Education, Hunan City University, Yiyang 413002, China
| | - Yushun Gong
- Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
36
|
Sgro M, Iacono G, Yamakawa GR, Kodila ZN, Marsland BJ, Mychasiuk R. Age matters: Microbiome depletion prior to repeat mild traumatic brain injury differentially alters microbial composition and function in adolescent and adult rats. PLoS One 2022; 17:e0278259. [PMID: 36449469 PMCID: PMC9710846 DOI: 10.1371/journal.pone.0278259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/13/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of the gut microbiome has been shown to perpetuate neuroinflammation, alter intestinal permeability, and modify repetitive mild traumatic brain injury (RmTBI)-induced deficits. However, there have been no investigations regarding the comparative effects that the microbiome may have on RmTBI in adolescents and adults. Therefore, we examined the influence of microbiome depletion prior to RmTBI on microbial composition and metabolome, in adolescent and adult Sprague Dawley rats. Rats were randomly assigned to standard or antibiotic drinking water for 14 days, and to subsequent sham or RmTBIs. The gut microbiome composition and metabolome were analysed at baseline, 1 day after the first mTBI, and at euthanasia (11 days following the third mTBI). At euthanasia, intestinal samples were also collected to quantify tight junction protein (TJP1 and occludin) expression. Adolescents were significantly more susceptible to microbiome depletion via antibiotic administration which increased pro-inflammatory composition and metabolites. Furthermore, RmTBI induced a transient increase in 'beneficial bacteria' (Lachnospiraceae and Faecalibaculum) in only adolescents that may indicate compensatory action in response to the injury. Finally, microbiome depletion prior to RmTBI generated a microbiome composition and metabolome that exemplified a potentially chronic pathogenic and inflammatory state as demonstrated by increased Clostridium innocuum and Erysipelatoclostridium and reductions in Bacteroides and Clostridium Sensu Stricto. Results highlight that adolescents are more vulnerable to RmTBI compared to adults and dysbiosis prior to injury may exacerbate secondary inflammatory cascades.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Giulia Iacono
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Zoe N. Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J. Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
37
|
Caffeine-Induced Sleep Restriction Alters the Gut Microbiome and Fecal Metabolic Profiles in Mice. Int J Mol Sci 2022; 23:ijms232314837. [PMID: 36499163 PMCID: PMC9737546 DOI: 10.3390/ijms232314837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Insufficient sleep is becoming increasingly common and contributes to many health issues. To combat sleepiness, caffeine is consumed daily worldwide. Thus, caffeine consumption and sleep restriction often occur in succession. The gut microbiome can be rapidly affected by either one's sleep status or caffeine intake, whereas the synergistic effects of a persistent caffeine-induced sleep restriction remain unclear. In this study, we investigated the impact of a chronic caffeine-induced sleep restriction on the gut microbiome and its metabolic profiles in mice. Our results revealed that the proportion of Firmicutes and Bacteroidetes was not altered, while the abundance of Proteobacteria and Actinobacteria was significantly decreased. In addition, the content of the lipids was abundant and significantly increased. A pathway analysis of the differential metabolites suggested that numerous metabolic pathways were affected, and the glycerophospholipid metabolism was most significantly altered. Combined analysis revealed that the metabolism was significantly affected by variations in the abundance and function of the intestinal microorganisms and was closely relevant to Proteobacteria and Actinobacteria. In conclusion, a long-term caffeine-induced sleep restriction affected the diversity and composition of the intestinal microbiota in mice, and substantially altered the metabolic profiles of the gut microbiome. This may represent a novel mechanism by which an unhealthy lifestyle such as mistimed coffee breaks lead to or exacerbates disease.
Collapse
|
38
|
Zhu H, Li G, Liu J, Xu X, Zhang Z. Gut microbiota is associated with the effect of photoperiod on seasonal breeding in male Brandt's voles (Lasiopodomys brandtii). MICROBIOME 2022; 10:194. [PMID: 36376894 PMCID: PMC9664686 DOI: 10.1186/s40168-022-01381-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/27/2022] [Indexed: 05/29/2023]
Abstract
BACKGROUND Seasonal breeding in mammals has been widely recognized to be regulated by photoperiod, but the association of gut microbiota with photoperiodic regulation of seasonal breeding has never been investigated. RESULTS In this study, we investigated the association of gut microbiota with photoperiod-induced reproduction in male Brandt's voles (Lasiopodomys brandtii) through a long-day and short-day photoperiod manipulation experiment and fecal microbiota transplantation (FMT) experiment. We found photoperiod significantly altered reproductive hormone and gene expression levels, and gut microbiota of voles. Specific gut microbes were significantly associated with the reproductive hormones and genes of voles during photoperiod acclimation. Transplantation of gut microbes into recipient voles induced similar changes in three hormones (melatonin, follicle-stimulating hormone, and luteinizing hormone) and three genes (hypothalamic Kiss-1, testicular Dio3, and Dio2/Dio3 ratio) to those in long-day and short-day photoperiod donor voles and altered circadian rhythm peaks of recipient voles. CONCLUSIONS Our study firstly revealed the association of gut microbiota with photoperiodic regulation of seasonal breeding through the HPG axis, melatonin, and Kisspeptin/GPR54 system. Our results may have significant implications for pest control, livestock animal breeding, and human health management. Video Abstract.
Collapse
Affiliation(s)
- Hanyi Zhu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoming Xu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
39
|
Gautier T, Olivieiro N, Ferron S, Le Pogam P, David-Le Gall S, Sauvager A, Leroyer P, Cannie I, Dion S, Sweidan A, Loréal O, Tomasi S, Bousarghin L. Bacteroides fragilis derived metabolites, identified by molecular networking, decrease Salmonella virulence in mice model. Front Microbiol 2022; 13:1023315. [DOI: 10.3389/fmicb.2022.1023315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
In the gut microbiota, resident bacteria prevent pathogens infection by producing specific metabolites. Among bacteria belonging to phylum Bacteroidota, we have previously shown that Bacteroides fragilis or its cell-free supernatant inhibited in vitro Salmonella Heidelberg translocation. In the present study, we have analyzed this supernatant to identify bioactive molecules after extraction and subsequent fractionation using a semi-preparative reversed-phase Liquid Chromatography High-Resolution Tandem Mass Spectrometry (LC-HRMS/MS). The results indicated that only two fractions (F3 and F4) strongly inhibited S. Heidelberg translocation in a model mimicking the intestinal epithelium. The efficiency of the bioactive fractions was evaluated in BALB/c mice, and the results showed a decrease of S. Heidelberg in Peyer’s patches and spleen, associated with a decrease in inflammatory cytokines and neutrophils infiltration. The reduction of the genus Alistipes in mice receiving the fractions could be related to the anti-inflammatory effects of bioactive fractions. Furthermore, these bioactive fractions did not alter the gut microbiota diversity in mice. To further characterize the compounds present in these bioactive fractions, Liquid Chromatography High-Resolution Tandem Mass Spectrometry (LC-HRMS/MS) data were analyzed through molecular networking, highlighting cholic acid (CA) and deoxycholic acid. In vitro, CA had inhibitory activity against the translocation of S. Heidelberg by significantly decreasing the expression of Salmonella virulence genes such as sipA. The bioactive fractions also significantly downregulated the flagellar gene fliC, suggesting the involvement of other active molecules. This study showed the interest to characterize better the metabolites produced by B. fragilis to make them means of fighting pathogenic bacteria by targeting their virulence factor without modifying the gut microbiota.
Collapse
|
40
|
Inoue M, Matsumura K, Sugimori N, Hamazaki K, Tsuchida A, Inadera H. Dietary intake of yogurt and cheese in children at age 1 year and sleep duration at age 1 and 3 years: the Japan Environment and Children's Study. BMC Pediatr 2022; 22:624. [PMID: 36319988 PMCID: PMC9623995 DOI: 10.1186/s12887-022-03633-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/15/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Background We previously reported that 1-year-old infants born to mothers who regularly consumed fermented food during pregnancy had a lower risk of sleep deprivation. However, it is not known if these positive effects are enhanced when infants themselves eat fermented foods or the long-term effects of such consumption. In this study, we examined the association between the frequency of fermented food intake during the child’s weaning period and sleep deprivation at age 1 and 3 years. Methods This birth cohort study used data from a nationwide, government-funded study called the Japan Environment and Children’s Study (JECS), covering 65,210 mother-child pairs. We examined the association between infants’ consumption of fermented foods at 1 year of age and sleep deprivation at 1 and 3 years of age. Results There was no association between yogurt or cheese intake and sleep duration at age 1; at age 3, there was no group difference, although a trend test showed that yogurt intake at age 1 was significantly associated with sleep duration at age 3. There was also no association between the frequency of cheese intake and inadequate sleep duration at age 3. Conclusion Frequency of children’s yogurt and cheese intake at age 1 was not associated with sleep duration at age 1 or 3. However, a trend test showed a significant association between the frequency of yogurt intake at age 1 and sleep duration at age 3.
Collapse
Affiliation(s)
- Mariko Inoue
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan
| | - Kenta Matsumura
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan ,grid.267346.20000 0001 2171 836XToyama Regional Center for JECS, University of Toyama, 2630 Sugitani, 930-8555 Toyama City, Toyama Japan
| | - Narumi Sugimori
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan
| | - Kei Hamazaki
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan ,grid.267346.20000 0001 2171 836XToyama Regional Center for JECS, University of Toyama, 2630 Sugitani, 930-8555 Toyama City, Toyama Japan ,grid.256642.10000 0000 9269 4097Department of Public Health, Gunma University Graduate School of Medicine, Showa 3-39-22, 371-8511 Maebashi, Gunma Japan
| | - Akiko Tsuchida
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan ,grid.267346.20000 0001 2171 836XToyama Regional Center for JECS, University of Toyama, 2630 Sugitani, 930-8555 Toyama City, Toyama Japan
| | - Hidekuni Inadera
- grid.267346.20000 0001 2171 836XDepartment of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, 930-0194 Toyama City, Toyama Japan ,grid.267346.20000 0001 2171 836XToyama Regional Center for JECS, University of Toyama, 2630 Sugitani, 930-8555 Toyama City, Toyama Japan
| | | |
Collapse
|
41
|
Legan TB, Lavoie B, Mawe GM. Direct and indirect mechanisms by which the gut microbiota influence host serotonin systems. Neurogastroenterol Motil 2022; 34:e14346. [PMID: 35246905 PMCID: PMC9441471 DOI: 10.1111/nmo.14346] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 12/18/2022]
Abstract
Mounting evidence highlights the pivotal role of enteric microbes as a dynamic interface with the host. Indeed, the gut microbiota, located in the lumen of the gastrointestinal (GI) tract, influence many essential physiological processes that are evident in both healthy and pathological states. A key signaling molecule throughout the body is serotonin (5-hydroxytryptamine; 5-HT), which acts in the GI tract to regulate numerous gut functions including intestinal motility and secretion. The gut microbiota can modulate host 5-HT systems both directly and indirectly. Direct actions of gut microbes, evidenced by studies using germ-free animals or antibiotic administration, alter the expression of key 5-HT-related genes to promote 5-HT biosynthesis. Indirectly, the gut microbiota produce numerous microbial metabolites, whose actions can influence host serotonergic systems in a variety of ways. This review summarizes the current knowledge regarding mechanisms by which gut bacteria act to regulate host 5-HT and 5-HT-mediated gut functions, as well as implications for 5-HT in the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Theresa B Legan
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Brigitte Lavoie
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Gary M Mawe
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
42
|
Chichlowski M, Cotter J, Fawkes N, Pandey N. Feed your microbiome and improve sleep, stress resilience, and cognition. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The brain and gut are connected both physically and biochemically. The gut-brain axis includes the central nervous system, neuroendocrine and neuroimmune systems, the enteric nervous system and vagus nerve, and the gut microbiome. It can influence brain function and even behavior, suggesting that dietary interventions may help enhance and protect mental health and cognitive performance. This review focuses on the role of the microbiome and its metabolites in sleep regulation, neurodegenerative disorders, mechanisms of stress, and mood. It also provides examples of nutritional interventions which can restore healthy gut microbiota and aid with risk reduction and management of many disorders related to mental and cognitive health. Evidence suggests a shift in the gut microbiota towards a balanced composition could be a target to maintain brain health, reduce stress and improve quality of life.
Collapse
Affiliation(s)
- Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN 47712, USA
| | - Jack Cotter
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| | - Neil Fawkes
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| | - Neeraj Pandey
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, SL1 3UH Slough, UK
| |
Collapse
|
43
|
Inoue M, Sugimori N, Hamazaki K, Matsumura K, Tsuchida A, Inadera H. Association between maternal fermented food consumption and child sleep duration at the age of 3 years: the Japan Environment and Children's Study. BMC Public Health 2022; 22:1504. [PMID: 35933371 PMCID: PMC9356427 DOI: 10.1186/s12889-022-13805-6] [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: 02/11/2022] [Accepted: 07/14/2022] [Indexed: 01/08/2023] Open
Abstract
Background Using cohort data from the Japan Environment and Children’s Study (JECS), we previously reported that the risk of sleep deprivation in 1-year-old children was reduced with a higher maternal intake of fermented foods, particularly miso. The present study, which evaluates children from the same cohort at 3 years of age, is a continuation of that work. Methods After applying exclusion criteria to 104,062 records in the JECS dataset, we evaluated 64,200 mother-child pairs in which the child was 3 years old. We examined the association of the dietary intake of fermented foods during pregnancy with child sleep duration < 10 h at the age of 3 years. Results Multivariable logistic regression analysis with the lowest quartile used as a reference revealed adjusted odds ratios (95% confidence intervals) for the second through fourth quartiles of 0.98 (0.90–1.06), 0.93 (0.85–1.01), and 0.85 (0.78–0.94) for cheese intake. Conclusions The consumption of fermented foods during pregnancy is associated with reduced risk of sleep deprivation in 3-year-old children, albeit in a limited way. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-022-13805-6.
Collapse
Affiliation(s)
- Mariko Inoue
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan
| | - Narumi Sugimori
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan
| | - Kei Hamazaki
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan.,Toyama Regional Center for Japan Environment and Children's Study, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.,Department of Public Health, Gunma University Graduate School of Medicine, Showa 3-39-22, Maebashi, Gunma, 371-8511, Japan
| | - Kenta Matsumura
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan.,Toyama Regional Center for Japan Environment and Children's Study, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Akiko Tsuchida
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan.,Toyama Regional Center for Japan Environment and Children's Study, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hidekuni Inadera
- Department of Public Health, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 30-0194, Japan. .,Toyama Regional Center for Japan Environment and Children's Study, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | | |
Collapse
|
44
|
The microbiota-gut-brain axis in sleep disorders. Sleep Med Rev 2022; 65:101691. [DOI: 10.1016/j.smrv.2022.101691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/04/2022] [Accepted: 08/19/2022] [Indexed: 12/25/2022]
|
45
|
Mediterranean Diet on Sleep: A Health Alliance. Nutrients 2022; 14:nu14142998. [PMID: 35889954 PMCID: PMC9318336 DOI: 10.3390/nu14142998] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 12/19/2022] Open
Abstract
The Mediterranean diet is a plant-based, antioxidant-rich, unsaturated fat dietary pattern that has been consistently associated with lower rates of noncommunicable diseases and total mortality, so that it is considered one of the healthiest dietary patterns. Clinical trials and mechanistic studies have demonstrated that the Mediterranean diet and its peculiar foods and nutrients exert beneficial effects against inflammation, oxidative stress, dysmetabolism, vascular dysfunction, adiposity, senescence, cognitive decline, neurodegeneration, and tumorigenesis, thus preventing age-associated chronic diseases and improving wellbeing and health. Nocturnal sleep is an essential physiological function, whose alteration is associated with health outcomes and chronic diseases. Scientific evidence suggests that diet and sleep are related in a bidirectional relationship, and the understanding of this association is important given their role in disease prevention. In this review, we surveyed the literature concerning the current state of evidence from epidemiological studies on the impact of the Mediterranean diet on nighttime sleep quantity and quality. The available studies indicate that greater adherence to the Mediterranean diet is associated with adequate sleep duration and with several indicators of better sleep quality. Potential mechanisms mediating the effect of the Mediterranean diet and its foods and nutrients on sleep are described, and gap-in-knowledge and new research agenda to corroborate findings are discussed.
Collapse
|
46
|
Zhang J, Zhang X, Zhang K, Lu X, Yuan G, Yang H, Guo H, Zhu Z, Wang T, Hao J, Sun Y, Su P, Zhang Z. The Component and Functional Pathways of Gut Microbiota Are Altered in Populations with Poor Sleep Quality - A Preliminary Report. Pol J Microbiol 2022; 71:241-250. [PMID: 35716170 PMCID: PMC9252145 DOI: 10.33073/pjm-2022-021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
With the development of genome sequencing, many researchers have investigated the mechanism by which the intestinal microbiota influences sleep across the brain-gut axis. However, the relationship between gut microbiota and sleep disorder remains unclear. Thus, we studied the difference in gut microbiota composition between poor sleep quality- and normal populations, which helps set the ground for future research. The recruited college students provided baseline information and stool samples and completed the Pittsburgh Sleep Quality Index (PSQI). We compared the two groups’ gut microbiota composition and functional differentiation by using the 16S rRNA gene sequencing analysis. The main bacterial difference and the most critical effect were mainly concentrated within Tenericutes and Elusimicrobia. Compared with the healthy control group, some functions of the gut microbiota were impaired in the poor sleep quality group, such as butanoate metabolism and propanoate metabolism. Bacterial taxa with significant differences raised the possibility for future diagnosis and treatment of sleep problems.
Collapse
Affiliation(s)
- Jianghui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Xueqing Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Kexin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Xiaoyan Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Guojing Yuan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Huayu Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Haiyun Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhihui Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Tianli Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jiahu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Ying Sun
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Puyu Su
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| |
Collapse
|
47
|
Zhang P, Kong L, Huang H, Pan Y, Zhang D, Jiang J, Shen Y, Xi C, Lai J, Ng CH, Hu S. Gut Microbiota – A Potential Contributor in the Pathogenesis of Bipolar Disorder. Front Neurosci 2022; 16:830748. [PMID: 35401095 PMCID: PMC8984199 DOI: 10.3389/fnins.2022.830748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/28/2022] [Indexed: 11/24/2022] Open
Abstract
Bipolar disorder (BD) is one of the major psychiatric disorders that is characterized by recurrent episodes of depression and mania (or hypomania), leading to seriously adverse outcomes with unclear pathogenesis. There is an underlying relationship between bacterial communities residing in the gut and brain function, which together form the gut-brain axis (GBA). Recent studies have shown that changes in the gut microbiota have been observed in a large number of BD patients, so the axis may play a role in the pathogenesis of BD. This review summarizes briefly the relationship between the GBA and brain function, the composition and changes of gut microbiota in patients with BD, and further explores the potential role of GBA-related pathway in the pathogenesis of BD as well as the limitations in this field at present in order to provide new ideas for the future etiology research and drug development.
Collapse
Affiliation(s)
- Peifen Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
| | - Lingzhuo Kong
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huimin Huang
- Department of Psychiatry, Wenzhou Medical University, Wenzhou, China
| | - Yanmeng Pan
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danhua Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajun Jiang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Shen
- Department of Psychiatry, Wenzhou Medical University, Wenzhou, China
| | - Caixi Xi
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
| | - Chee H. Ng
- Department of Psychiatry, The Melbourne Clinic and St Vincent’s Hospital, University of Melbourne, Richmond, VIC, Australia
- *Correspondence: Chee H. Ng,
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, Hangzhou, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Shaohua Hu,
| |
Collapse
|
48
|
Akagawa S, Kaneko K. Gut microbiota and allergic diseases in children. Allergol Int 2022; 71:301-309. [PMID: 35314107 DOI: 10.1016/j.alit.2022.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota resides in the human gastrointestinal tract, where it plays an important role in maintaining host health. The human gut microbiota is established by the age of 3 years. Studies have revealed that an imbalance in the gut microbiota, termed dysbiosis, occurs due to factors such as cesarean delivery and antibiotic use before the age of 3 years and that dysbiosis is associated with a higher risk of future onset of allergic diseases. Recent advancements in next-generation sequencing methods have revealed the presence of dysbiosis in patients with allergic diseases, which increases attention on the relationship between dysbiosis and the development of allergic diseases. However, there is no unified perspective on the characteristics on dysbiosis or the mechanistic link between dysbiosis and the onset of allergic diseases. Here, we introduce the latest studies on the gut microbiota in children with allergic diseases and present the hypothesis that dysbiosis characterized by fewer butyric acid-producing bacteria leads to fewer regulatory T cells, resulting in allergic disease. Further studies on correcting dysbiosis for the prevention and treatment of allergic diseases are warranted.
Collapse
Affiliation(s)
- Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Osaka, Japan.
| |
Collapse
|
49
|
One Giant Leap from Mouse to Man: The Microbiota-Gut-Brain Axis in Mood Disorders and Translational Challenges Moving towards Human Clinical Trials. Nutrients 2022; 14:nu14030568. [PMID: 35276927 PMCID: PMC8840472 DOI: 10.3390/nu14030568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
The microbiota–gut–brain axis is a bidirectional communication pathway that enables the gut microbiota to communicate with the brain through direct and indirect signaling pathways to influence brain physiology, function, and even behavior. Research has shown that probiotics can improve several aspects of health by changing the environment within the gut, and several lines of evidence now indicate a beneficial effect of probiotics on mental and brain health. Such evidence has prompted the arrival of a new term to the world of biotics research: psychobiotics, defined as any exogenous influence whose effect on mental health is bacterially mediated. Several taxonomic changes in the gut microbiota have been reported in neurodevelopmental disorders, mood disorders such as anxiety and depression, and neurodegenerative disorders such as Alzheimer’s disease. While clinical evidence supporting the role of the gut microbiota in mental and brain health, and indeed demonstrating the beneficial effects of probiotics is rapidly accumulating, most of the evidence to date has emerged from preclinical studies employing different animal models. The purpose of this review is to focus on the role of probiotics and the microbiota–gut–brain axis in relation to mood disorders and to review the current translational challenges from preclinical to clinical research.
Collapse
|
50
|
Suda K, Matsuda K. How Microbes Affect Depression: Underlying Mechanisms via the Gut-Brain Axis and the Modulating Role of Probiotics. Int J Mol Sci 2022; 23:ijms23031172. [PMID: 35163104 PMCID: PMC8835211 DOI: 10.3390/ijms23031172] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Accumulating evidence suggests that the gut microbiome influences the brain functions and psychological state of its host via the gut-brain axis, and gut dysbiosis has been linked to several mental illnesses, including major depressive disorder (MDD). Animal experiments have shown that a depletion of the gut microbiota leads to behavioral changes, and is associated with pathological changes, including abnormal stress response and impaired adult neurogenesis. Short-chain fatty acids such as butyrate are known to contribute to the up-regulation of brain-derived neurotrophic factor (BDNF), and gut dysbiosis causes decreased levels of BDNF, which could affect neuronal development and synaptic plasticity. Increased gut permeability causes an influx of gut microbial components such as lipopolysaccharides, and the resultant systemic inflammation may lead to neuroinflammation in the central nervous system. In light of the fact that gut microbial factors contribute to the initiation and exacerbation of depressive symptoms, this review summarizes the current understanding of the molecular mechanisms involved in MDD onset, and discusses the therapeutic potential of probiotics, including butyrate-producing bacteria, which can mediate the microbiota-gut-brain axis.
Collapse
|