1
|
Singh A, Negi PS. Appraising the role of biotics and fermented foods in gut microbiota modulation and sleep regulation. J Food Sci 2025. [PMID: 39750017 DOI: 10.1111/1750-3841.17634] [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: 09/10/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
Sleep disturbances are increasingly prevalent, significantly impacting physical and mental health. Recent research reveals a bidirectional relationship between gut microbiota and sleep, mediated through the microbiota-gut-brain axis. This review examines the role of gut microbiota in sleep physiology and explores how biotics, including probiotics, prebiotics, synbiotics, postbiotics, and fermented foods, can enhance sleep quality. Drawing from animal and human studies, we discuss neurobiological mechanisms by which biotics may influence sleep, including modulation of neurotransmitters, immune responses, and hormonal regulation. Key microbial metabolites, such as short-chain fatty acids, are highlighted for their role in supporting sleep-related neurochemical processes. Additionally, this review presents dietary strategies and food processing technologies, like fermentation, as innovative approaches for sleep enhancement. Although promising, the available research has limitations, including small sample sizes, variability in biotic strains and dosages, and reliance on subjective sleep assessments. This review underscores the need for standardized protocols, objective assessments such as polysomnography, and personalized biotic interventions. Emerging findings highlight the therapeutic potential of gut microbiota modulation for sleep improvement, though further large-scale human trials are essential to refine strain selection, dosage, and formulation. This interdisciplinary exploration seeks to advance food-based interventions and holistic strategies for managing sleep disorders and improving quality of life.
Collapse
Affiliation(s)
- Akanksha Singh
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Pradeep Singh Negi
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| |
Collapse
|
2
|
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
|
3
|
Murack M, Kadamani AK, Guindon-Riopel A, Traynor OH, Iqbal UH, Bronner S, Messier C, Ismail N. The effect of probiotic supplementation on sleep, depression-like behaviour, and central glucose and lactate metabolism in male and female pubertal mice exposed to chronic sleep disruption. Psychoneuroendocrinology 2024; 168:107146. [PMID: 39079447 DOI: 10.1016/j.psyneuen.2024.107146] [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: 03/24/2024] [Revised: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024]
Abstract
The prevalence of depression significantly increases during puberty and adolescence. Puberty is the period during which sexual maturity is attained, while adolescence persists beyond puberty and includes physiological, social, emotional, and cognitive maturation. A stressor that has been shown previously to induce depression is chronic sleep disruption. Probiotics can prevent stress-induced depression. However, it was unclear whether probiotics could prevent depression following chronic sleep disruption and what mechanism may be involved. Therefore, we investigated whether pubertal probiotic treatment could prevent depression-like behavior in mice following chronic sleep disruption. We also examined whether probiotic treatment could improve sleep quality, and increase serotonin, tryptophan, glucose, and L-lactate concentrations in chronically sleep-disrupted mice. We hypothesized that probiotic treatment would prevent depression-like behavior, improve sleep quality, and increase serotonin, tryptophan, glucose, and L-lactate concentrations in sleep-disrupted mice. Male and female mice (N=120) received cannula and electroencephalogram (EEG) electrode implants at postnatal day (PND) 26. Mice received Lacidofil® or Cerebiome® probiotics (PND 33-51) and were sleep-disrupted for the first 4 hours of the light phase (sleep period) (PND 40-51). Hippocampal L-lactate and glucose concentrations and sleep were measured over a 24-h period (PND 48-49). Depression-like behaviour was evaluated using tail suspension (PND 49) and forced swim tests (PND 50). Chronic sleep disruption increased depression-like behaviour and NREM duration in the dark phase, and reduced all metabolites and neuromodulating biomolecules measured within the brain. However, mice treated with probiotics did not display depression-like behaviour or decreased hippocampal L-lactate following chronic sleep disruption. Cerebiome prevented decreases to prefrontal serotonin and hippocampal glucose concentrations, while Lacidofil increased NREM duration in the latter half of the light phase. The current study not only replicates previous findings linking chronic sleep disruption to depression, but also demonstrates that pubertal probiotic treatment can mitigate the effects of chronic sleep disruption on depression-like behaviour and on the neural mechanisms underlying depression in a strain-dependent manner.
Collapse
Affiliation(s)
- Michael Murack
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada
| | - Anthony K Kadamani
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada
| | - Alexi Guindon-Riopel
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada
| | - Olivia H Traynor
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada
| | - Umar Haris Iqbal
- Rosell Institute for Microbiome and Probiotics, 6100 Royalmont Avenue, Montreal, Quebec H4P 2R2, Canada
| | - Stéphane Bronner
- Rosell Institute for Microbiome and Probiotics, 6100 Royalmont Avenue, Montreal, Quebec H4P 2R2, Canada
| | - Claude Messier
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, University of Ottawa, 136 Jean-Jacques Lussier, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
4
|
Slykerman R, Davies N, Fuad M, Dekker J. Milk fat globule membranes for psychological and physical health: qualitative results from the Employing Milk Phospholipids to Observe Well-being and Emotional Resilience (EMPOWER) randomised trial. J Hum Nutr Diet 2024; 37:1091-1099. [PMID: 38798237 DOI: 10.1111/jhn.13326] [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: 01/14/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Milk fat globule membranes (MFGM) present a nutritional intervention with the potential to improve psychological well-being and mitigate the negative effects of stress on health. The present study aimed to investigate participant's experience of different aspects of health during a trial of MFGM supplementation and determine the effect of MFGM on qualitative measures of psychological and physical well-being. METHODS Seventy-three adults in New Zealand who were enrolled in a clinical trial to test MFGM supplementation for improvement of psychological well-being took part in a post-intervention interview. Participants and researchers remained blinded to intervention group allocation. Interviews were conducted over the video conferencing platform Zoom and transcribed. A mixed methods analytical approach included thematic analysis to identify emerging themes and χ2 regression models to examine frequency of improvements in different aspects of well-being between the MFGM and placebo groups. RESULTS There were no significant demographic or psychological differences between interviewees and non-interviewed study participants. Four central themes emerged from the data for all participants: improved well-being, increased ability to cope with stress and improvements in mood, improvement in physical energy or activity, and improved sleep. The frequency of improved ability to cope with stress and improved sleep quality was significantly higher in participants who received MFGM supplementation compared to those receiving the placebo. CONCLUSIONS Qualitative data may capture aspects of improved sleep or psychological well-being not measured by rating scales. The results suggest that MFGM supplementation may improve the ability to cope with stress and improve sleep quality in healthy adults.
Collapse
Affiliation(s)
- Rebecca Slykerman
- Department of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| | - Naomi Davies
- Department of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| | - Maher Fuad
- Fonterra Research and Development Centre, Fonterra Cooperative Group Limited, Palmerston North, New Zealand
| | - James Dekker
- Fonterra Research and Development Centre, Fonterra Cooperative Group Limited, Palmerston North, New Zealand
| |
Collapse
|
5
|
Morales-Suárez-Varela M, Amezcua-Prieto C, Peraita-Costa I, Mateos-Campos R, Ayán C, Ortiz-Moncada R, Fernández-Villa T. Sleep Patterns and Tryptophan Consumption among Students at Spanish Universities: The Unihcos Project. Nutrients 2024; 16:2376. [PMID: 39064819 PMCID: PMC11279726 DOI: 10.3390/nu16142376] [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: 07/10/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The objective of this cross-sectional study was to explore sleep patterns and the potential relationship between sleep and tryptophan intake among Spanish university students. A total of 11,485 students self-reported their sleep and dietary patterns and habits. Tryptophan intake was calculated using a food intake matrix and results were presented as quartiles of total intake. Short sleep duration prevalence was 51.0%, with males exhibiting a significantly higher frequency. A total of 55.0% of participants presented inadequate sleep efficiency, with males again presenting a higher rate. Median tryptophan intake was 692.16 ± 246.61 mg/day, 731.84 ± 246.86 mg/day in males and 677.24 ± 244.87 mg/day in females (p = 0.001). Dietary tryptophan intake below the first quartile (<526.43 mg/day) was associated with a higher risk of short sleep duration in males (1.26; 95%CI: 1.02-1.55) and females (1.19; 95%CI: 1.05-1.34) and with the Athens Insomnia Scale insomnia in males (2.56; 95%CI: 1.36-4.82) and females (1.47; 95%CI: 1.10-2.05). Regarding academic specializations, females in the humanities field showed a higher risk of Athens Insomnia Scale insomnia due to low tryptophan intake (Q1: 3.15; 95% CI: 1.04-9.55 and Q2: 3.41; 95%CI: 1.01-11.5). In summary, lower tryptophan consumption appears to be associated with poorer sleep quality in Spanish university students; however, other social factors affecting students may also influence sleep quality. These findings have important implications for nutritional recommendations aimed at enhancing tryptophan intake to improve sleep quality.
Collapse
Affiliation(s)
- María Morales-Suárez-Varela
- Research Group in Social and Nutritional Epidemiology, Pharmacoepidemiology and Public Health, Department of Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine, Faculty of Pharmacy and Food Sciences, Universitat de València, Av. Vicent Andrés Estelles s/n, 46100 Burjassot, Spain;
- Biomedical Research Center in Epidemiology and Public Health Network (CIBERESP), Carlos III Health Institute, Av. Monforte de Lemos 3-5 Pabellón 11 Planta 0, 28029 Madrid, Spain; (C.A.-P.); (T.F.-V.)
| | - Carmen Amezcua-Prieto
- Biomedical Research Center in Epidemiology and Public Health Network (CIBERESP), Carlos III Health Institute, Av. Monforte de Lemos 3-5 Pabellón 11 Planta 0, 28029 Madrid, Spain; (C.A.-P.); (T.F.-V.)
- Department of Preventive Medicine and Public Health, Universidad de Granada, Avenida de la Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de Madrid, 15, 18071 Granada, Spain
| | - Isabel Peraita-Costa
- Research Group in Social and Nutritional Epidemiology, Pharmacoepidemiology and Public Health, Department of Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine, Faculty of Pharmacy and Food Sciences, Universitat de València, Av. Vicent Andrés Estelles s/n, 46100 Burjassot, Spain;
- Biomedical Research Center in Epidemiology and Public Health Network (CIBERESP), Carlos III Health Institute, Av. Monforte de Lemos 3-5 Pabellón 11 Planta 0, 28029 Madrid, Spain; (C.A.-P.); (T.F.-V.)
| | - Ramona Mateos-Campos
- Area of Preventive Medicine and Public Health, Department of Biomedical and Diagnostic Sciences, Universidad de Salamanca, Calle Alfonso X el Sabio s/n, 37007 Salamanca, Spain;
| | - Carlos Ayán
- School of Education and Sports Sciences, HealthyFit Research Group, Universidad de Vigo, Campus A Xunqueira s/n, 36005 Pontevedra, Spain;
| | - Rocío Ortiz-Moncada
- Area of Preventive Medicine and Public Health, Department of Community Nursing, Preventive Medicine and Public Health and History of Science, Food and Nutrition Research Group, Universidad de Alicante, Carretera de San Vicente del Raspeig s/n, San Vicente del Raspeig, 03690 Alicante, Spain;
| | - Tania Fernández-Villa
- Biomedical Research Center in Epidemiology and Public Health Network (CIBERESP), Carlos III Health Institute, Av. Monforte de Lemos 3-5 Pabellón 11 Planta 0, 28029 Madrid, Spain; (C.A.-P.); (T.F.-V.)
- Department of Biomedical Sciences, Area of Preventive Medicine and Public Health, Universidad de León, Campus Universitario de Vegazana, 24071 León, Spain
- Group of Investigation in Interactions Gene-Environment and Health (GIIGAS), Institute of Biomedicine (IBIOMED), Universidad de León, Campus Universitario de Vegazana, 24071 León, Spain
| |
Collapse
|
6
|
Lin Z, Jiang T, Chen M, Ji X, Wang Y. Gut microbiota and sleep: Interaction mechanisms and therapeutic prospects. Open Life Sci 2024; 19:20220910. [PMID: 39035457 PMCID: PMC11260001 DOI: 10.1515/biol-2022-0910] [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: 03/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024] Open
Abstract
Sleep is crucial for wellness, and emerging research reveals a profound connection to gut microbiota. This review explores the bidirectional relationship between gut microbiota and sleep, exploring the mechanisms involved and the therapeutic opportunities it presents. The gut-brain axis serves as a conduit for the crosstalk between gut microbiota and the central nervous system, with dysbiosis in the microbiota impairing sleep quality and vice versa. Diet, circadian rhythms, and immune modulation all play a part. Specific gut bacteria, like Lactobacillus and Bifidobacterium, enhance sleep through serotonin and gamma-aminobutyric acid production, exemplifying direct microbiome influence. Conversely, sleep deprivation reduces beneficial bacteria, exacerbating dysbiosis. Probiotics, prebiotics, postbiotics, and fecal transplants show therapeutic potential, backed by animal and human research, yet require further study on safety and long-term effects. Unraveling this intricate link paves the way for tailored sleep therapies, utilizing microbiome manipulation to improve sleep and health. Accelerated research is essential to fully tap into this promising field for sleep disorder management.
Collapse
Affiliation(s)
- Zhonghui Lin
- Department of Neurology Medical, Xiamen Hospital of Traditional Chinese Medicine, Fujian, Xiamen, China
- Jimsar County of Xinjiang Chinese Medicine Hospital, Xinjiang, Changji, China
| | - Tao Jiang
- Department of Neurology Medical, Xiamen Hospital of Traditional Chinese Medicine, Fujian, Xiamen, China
| | - Miaoling Chen
- Department of Neurology Medical, Xiamen Hospital of Traditional Chinese Medicine, Fujian, Xiamen, China
| | - Xudong Ji
- Jimsar County of Xinjiang Chinese Medicine Hospital, Xinjiang, Changji, China
| | - Yunsu Wang
- Department of Cardiology Medical, Xiamen Hospital of Traditional Chinese Medicine, Fujian, Xiamen, China
| |
Collapse
|
7
|
Thompson RS, Bowers SJ, Vargas F, Hopkins S, Kelley T, Gonzalez A, Lowry CA, Dorrestein PC, Vitaterna MH, Turek FW, Knight R, Wright KP, Fleshner M. A Prebiotic Diet Containing Galactooligosaccharides and Polydextrose Produces Dynamic and Reproducible Changes in the Gut Microbial Ecosystem in Male Rats. Nutrients 2024; 16:1790. [PMID: 38892722 PMCID: PMC11175065 DOI: 10.3390/nu16111790] [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: 05/07/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Despite substantial evidence supporting the efficacy of prebiotics for promoting host health and stress resilience, few experiments present evidence documenting the dynamic changes in microbial ecology and fecal microbially modified metabolites over time. Furthermore, the literature reports a lack of reproducible effects of prebiotics on specific bacteria and bacterial-modified metabolites. The current experiments examined whether consumption of diets enriched in prebiotics (galactooligosaccharides (GOS) and polydextrose (PDX)), compared to a control diet, would consistently impact the gut microbiome and microbially modified bile acids over time and between two research sites. Male Sprague Dawley rats were fed control or prebiotic diets for several weeks, and their gut microbiomes and metabolomes were examined using 16S rRNA gene sequencing and untargeted LC-MS/MS analysis. Dietary prebiotics altered the beta diversity, relative abundance of bacterial genera, and microbially modified bile acids over time. PICRUSt2 analyses identified four inferred functional metabolic pathways modified by the prebiotic diet. Correlational network analyses between inferred metabolic pathways and microbially modified bile acids revealed deoxycholic acid as a potential network hub. All these reported effects were consistent between the two research sites, supporting the conclusion that dietary prebiotics robustly changed the gut microbial ecosystem. Consistent with our previous work demonstrating that GOS/PDX reduces the negative impacts of stressor exposure, we propose that ingesting a diet enriched in prebiotics facilitates the development of a health-promoting gut microbial ecosystem.
Collapse
Affiliation(s)
- Robert S. Thompson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Samuel J. Bowers
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL 60208, USA; (S.J.B.); (M.H.V.); (F.W.T.)
| | - Fernando Vargas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA 92093, USA (P.C.D.)
| | - Shelby Hopkins
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Tel Kelley
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA (R.K.)
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA 92093, USA (P.C.D.)
| | - Martha Hotz Vitaterna
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL 60208, USA; (S.J.B.); (M.H.V.); (F.W.T.)
| | - Fred W. Turek
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL 60208, USA; (S.J.B.); (M.H.V.); (F.W.T.)
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA (R.K.)
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA 92093, USA
- Center for Microbiome Innovation, University of California San Diego, San Diego, CA 92093, USA
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (R.S.T.); (T.K.); (C.A.L.); (K.P.W.J.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
8
|
Deyang T, Baig MAI, Dolkar P, Hediyal TA, Rathipriya AG, Bhaskaran M, PandiPerumal SR, Monaghan TM, Mahalakshmi AM, Chidambaram SB. Sleep apnoea, gut dysbiosis and cognitive dysfunction. FEBS J 2024; 291:2519-2544. [PMID: 37712936 DOI: 10.1111/febs.16960] [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: 05/26/2023] [Revised: 08/14/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
Sleep disorders are becoming increasingly common, and their distinct effects on physical and mental health require elaborate investigation. Gut dysbiosis (GD) has been reported in sleep-related disorders, but sleep apnoea is of particular significance because of its higher prevalence and chronicity. Cumulative evidence has suggested a link between sleep apnoea and GD. This review highlights the gut-brain communication axis that is mediated via commensal microbes and various microbiota-derived metabolites (e.g. short-chain fatty acids, lipopolysaccharide and trimethyl amine N-oxide), neurotransmitters (e.g. γ-aminobutyric acid, serotonin, glutamate and dopamine), immune cells and inflammatory mediators, as well as the vagus nerve and hypothalamic-pituitary-adrenal axis. This review also discusses the pathological role underpinning GD and altered gut bacterial populations in sleep apnoea and its related comorbid conditions, particularly cognitive dysfunction. In addition, the review examines the preclinical and clinical evidence, which suggests that prebiotics and probiotics may potentially be beneficial in sleep apnoea and its comorbidities through restoration of eubiosis or gut microbial homeostasis that regulates neural, metabolic and immune responses, as well as physiological barrier integrity via the gut-brain axis.
Collapse
Affiliation(s)
- Tenzin Deyang
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Md Awaise Iqbal Baig
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Phurbu Dolkar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, Frederic and Mary Wolf Center, University of Toledo Health Science Campus, OH, USA
| | - Seithikuruppu R PandiPerumal
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Division of Research and Development, Lovely Professional University, Phagwara, India
| | - Tanya M Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, UK
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| |
Collapse
|
9
|
Freitas SM, Franco B, Saragiotto G, Morais MA, Simabuco FM, Cunha DT, Esteves AM, Antunes AEC. Effect of a probiotic fermented milk supplementation on behavior and sleep. Nutr Neurosci 2024; 27:607-619. [PMID: 37496309 DOI: 10.1080/1028415x.2023.2240990] [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: 07/28/2023]
Abstract
This study attempted to analyze the effect of supplementing Wistar-Kyoto rats with fermented milk containing the probiotic Bifidobacterium animalis BB-12 and pomegranate juice on the microbiota-gut-brain axis of rats, with special focus on their behavior, sleep patterns, and response to stress. This study was divided into two experiments: (1) For the behavioral analysis the animals were divided into two groups: Fermented probiotic milk (BB + 1) and control (BB-). (2) For the sleep analysis the animals were divided into two groups: Fermented probiotic milk (BB + 2) and control (H2O). For the behavioral analysis, the open field method was used, which evaluates the behavior after ten, twenty, and thirty days of supplementation. For sleep analysis, the animals were submitted to implantation of electrodes and 24 h polysomnography, followed by 48 h sleep deprivation (REM) and 48 h polysomnography, then euthanized 100 days after the beginning of the experiment. In addition, animal feces were collected before and after sleep deprivation to assess its effects on the microbiota. A decrease in anxiety-related behaviors was observed in the supplemented animals and an increase in sleep efficiency and a reduction in the number of awakenings of the animals before deprivation. It has also been observed that sleep deprivation decreased the amount of total bacterial DNA. The number of copies of genomes of the genus Bifidobacterium did not differ in both groups.
Collapse
Affiliation(s)
- Samara M Freitas
- School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Beatriz Franco
- School of Physical Education, University of Campinas, Campinas, Brazil
| | | | - Milca A Morais
- School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Diogo T Cunha
- School of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Andrea M Esteves
- School of Applied Sciences, University of Campinas, Limeira, Brazil
- School of Physical Education, University of Campinas, Campinas, Brazil
| | | |
Collapse
|
10
|
Wang Y, Du W, Hu X, Yu X, Guo C, Jin X, Wang W. Targeting the blood-brain barrier to delay aging-accompanied neurological diseases by modulating gut microbiota, circadian rhythms, and their interplays. Acta Pharm Sin B 2023; 13:4667-4687. [PMID: 38045038 PMCID: PMC10692395 DOI: 10.1016/j.apsb.2023.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/05/2023] [Accepted: 08/02/2023] [Indexed: 12/05/2023] Open
Abstract
The blood-brain barrier (BBB) impairment plays a crucial role in the pathological processes of aging-accompanied neurological diseases (AAND). Meanwhile, circadian rhythms disruption and gut microbiota dysbiosis are associated with increased morbidity of neurological diseases in the accelerated aging population. Importantly, circadian rhythms disruption and gut microbiota dysbiosis are also known to induce the generation of toxic metabolites and pro-inflammatory cytokines, resulting in disruption of BBB integrity. Collectively, this provides a new perspective for exploring the relationship among circadian rhythms, gut microbes, and the BBB in aging-accompanied neurological diseases. In this review, we focus on recent advances in the interplay between circadian rhythm disturbances and gut microbiota dysbiosis, and their potential roles in the BBB disruption that occurs in AAND. Based on existing literature, we discuss and propose potential mechanisms underlying BBB damage induced by dysregulated circadian rhythms and gut microbiota, which would serve as the basis for developing potential interventions to protect the BBB in the aging population through targeting the BBB by exploiting its links with gut microbiota and circadian rhythms for treating AAND.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurology, the Second Affiliated Hospital of Jiaxing City, Jiaxing 314000, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xin Yu
- Bengbu Medical College (Department of Neurology, the Second Hospital of Jiaxing City), Jiaxing 233030, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Wei Wang
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| |
Collapse
|
11
|
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
|
12
|
Cui S, Xiao H, Miao D, Yang W. Metal uptake and translocation by Chinese brake fern (Pteris vittata) and diversity of rhizosphere microbial communities under single and combined arsenic and cadmium stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:85198-85209. [PMID: 37380855 DOI: 10.1007/s11356-023-28448-2] [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: 02/07/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023]
Abstract
Chinese brake fern (Pteris vittata) can increase tolerance to arsenic (As) and cadmium (Cd) toxicity by regulating rhizosphere microbial diversity. However, effects of combined As-Cd stress on microbial diversity and plant uptake and transport remain poorly understood. Therefore, effects of different concentrations of As and Cd on Pteris vittata (P. vittata) metal uptake and translocation and rhizosphere microbial diversity were examined in a pot experiment. The results indicated that As primarily accumulated aboveground in P. vittata (bioconcentration factor (BCF) ≤ 51.3; translocation factor (TF) ≈ 4), whereas Cd primarily accumulated belowground (BCF ≤ 39.1; TF < 1). Under single As, single Cd, and As-Cd combined stress, the most dominant bacteria and fungi were Burkholderia-Caballeronia-P (6.62-27.92%) and Boeremia (4.61-30.42%), Massilia (8.07-11.51%) and Trichoderma (4.47-22.20%), and Bradyrhizobium (2.24-10.38%) and Boeremia (3.16-45.69%), respectively, and their abundance ratios had a significant impact on the efficiency of P. vittata for As and Cd accumulation. However, with increasing As and Cd concentrations, abundances of plant pathogenic bacteria such as Fusarium and Chaetomium (the highest abundances were 18.08% and 23.72%, respectively) increased, indicating that As and Cd concentrations reduced P. vittata resistance to pathogens. At high soil concentrations of As-Cd, although plant As and Cd contents increased and microbial diversity was highest, enrichment efficiency and transportability of As and Cd decreased substantially. Therefore, pollution intensity should be considered when evaluating P. vittata suitability for phytoremediation of combined As-Cd contaminated soils.
Collapse
Affiliation(s)
- Shizhan Cui
- School of Agriculture and Life Sciences, Kunming University, Kunming, 650214, Yunnan, China
| | - Han Xiao
- School of Chemistry and Chemical Engineering, Kunming University, Kunming, 650214, Yunnan, China
| | - Deren Miao
- School of Chemistry and Chemical Engineering, Kunming University, Kunming, 650214, Yunnan, China
| | - Wanqiu Yang
- School of Chemistry and Chemical Engineering, Kunming University, Kunming, 650214, Yunnan, China.
| |
Collapse
|
13
|
Li L, Liang T, Jiang T, Li Y, Yang L, Wu L, Yang J, Ding Y, Wang J, Chen M, Zhang J, Xie X, Wu Q. Gut microbiota: Candidates for a novel strategy for ameliorating sleep disorders. Crit Rev Food Sci Nutr 2023; 64:10772-10788. [PMID: 37477274 DOI: 10.1080/10408398.2023.2228409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The aim of this review was to evaluate the feasibility of treating sleep disorders using novel gut microbiota intervention strategies. Multiple factors can cause sleep disorders, including an imbalance in the gut microbiota. Studies of the microbiome-gut-brain axis have revealed bidirectional communication between the central nervous system and gut microbes, providing a more comprehensive understanding of mood and behavioral regulatory patterns. Changes in the gut microbiota and its metabolites can stimulate the endocrine, nervous, and immune systems, which regulate the release of neurotransmitters and alter the activity of the central nervous system, ultimately leading to sleep disorders. Here, we review the main factors affecting sleep, discuss possible pathways and molecular mechanisms of the interaction between sleep and the gut microbiota, and compare common gut microbiota intervention strategies aimed at improving sleep physiology.
Collapse
Affiliation(s)
- Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Tingting Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Tong Jiang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Lingshuang Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Lei Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Juan Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
14
|
Bester A, O'Brien M, Cotter PD, Dam S, Civai C. Shotgun Metagenomic Sequencing Revealed the Prebiotic Potential of a Fruit Juice Drink with Fermentable Fibres in Healthy Humans. Foods 2023; 12:2480. [PMID: 37444219 DOI: 10.3390/foods12132480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Fibre-based dietary interventions are at the forefront of gut microbiome modulation research, with a wealth of 16S rRNA information to demonstrate the prebiotic effects of isolated fibres. However, there is a distinct lack of data relating to the effect of a combination of soluble and insoluble fibres in a convenient-to-consume fruit juice food matrix on gut microbiota structure, diversity, and function. Here, we aimed to determine the impact of the MOJU Prebiotic Shot, an apple, lemon, ginger, and raspberry fruit juice drink blend containing chicory inulin, baobab, golden kiwi, and green banana powders, on gut microbiota structure and function. Healthy adults (n = 20) were included in a randomised, double-blind, placebo-controlled, cross-over study, receiving 60 mL MOJU Prebiotic Shot or placebo (without the fibre mix) for 3 weeks with a 3-week washout period between interventions. Shotgun metagenomics revealed significant between-group differences in alpha and beta diversity. In addition, the relative abundance of the phyla Actinobacteria and Desulfobacteria was significantly increased as a result of the prebiotic intervention. Nine species were observed to be differentially abundant (uncorrected p-value of <0.05) as a result of the prebiotic treatment. Of these, Bifidobacterium adolescentis and CAG-81 sp900066785 (Lachnospiraceae) were present at increased abundance relative to baseline. Additionally, KEGG analysis showed an increased abundance in pathways associated with arginine biosynthesis and phenylacetate degradation during the prebiotic treatment. Our results show the effects of the daily consumption of 60 mL MOJU Prebiotic Shot for 3 weeks and provide insight into the functional potential of B. adolescentis.
Collapse
Affiliation(s)
- Adri Bester
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| | | | | | | | - Claudia Civai
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| |
Collapse
|
15
|
Hopkins S, Kelley T, Roller R, Thompson RS, Colagiovanni DB, Chupka K, Fleshner M. Oral CBD-rich hemp extract modulates sterile inflammation in female and male rats. Front Physiol 2023; 14:1112906. [PMID: 37275221 PMCID: PMC10234154 DOI: 10.3389/fphys.2023.1112906] [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: 11/30/2022] [Accepted: 04/26/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction: Cannabidiol (CBD) extract from the cannabis plant has biomedical and nutraceutical potential. Unlike tetrahydrocannabinol (THC), CBD products produce few psychoactive effects and pose little risk for abuse. There is emerging preclinical and clinical evidence that CBD is stress modulatory and may have anti-inflammatory properties. People across the United States legally ingest CBD-rich hemp extracts to manage mental and physical health problems, including stress and inflammation. Preclinical studies have revealed potential mechanisms for these effects; however, the impact of this prior work is diminished because many studies: 1) tested synthetic CBD rather than CBD-rich hemp extracts containing terpenes and/or other cannabinoids thought to enhance therapeutic benefits; 2) administered CBD via injection into the peritoneal cavity or the brain instead of oral ingestion; and 3) failed to examine potential sex differences. To address these gaps in the literature, the following study tested the hypothesis that the voluntary oral ingestion of CBD-rich hemp extract will attenuate the impact of stressor exposure on plasma and tissue inflammatory and stress proteins in females and males. Methods: Adult male and female Sprague Dawley rats (10-15/group) were randomly assigned to be given cereal coated with either vehicle (coconut oil) or CBD-rich hemp extract (L-M0717, CBDrx/Functional Remedies, 20.0 mg/kg). After 7 days, rats were exposed to a well-established acute model of stress (100, 1.5 mA, 5-s, intermittent tail shocks, 90 min total duration) or remained in home cages as non-stressed controls. Results: Stressor exposure induced a robust stress response, i.e., increased plasma corticosterone and blood glucose, and decreased spleen weight (a surrogate measure of sympathetic nervous system activation). Overall, stress-induced increases in inflammatory and stress proteins were lower in females than males, and oral CBD-rich hemp extract constrained these responses in adipose tissue (AT) and mesenteric lymph nodes (MLN). Consistent with previous reports, females had higher levels of stress-evoked corticosterone compared to males, which may have contributed to the constrained inflammatory response measured in females. Discussion: Results from this study suggest that features of the acute stress response are impacted by oral ingestion of CBD-rich hemp extract in female and male rats, and the pattern of changes may be sex and tissue dependent.
Collapse
Affiliation(s)
- Shelby Hopkins
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| | - Tel Kelley
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
| | - Rachel Roller
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
| | - Robert S. Thompson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| | | | - Kris Chupka
- Next Frontier Biosciences, Westminster, CO, United States
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, United States
| |
Collapse
|
16
|
Freijy TM, Cribb L, Oliver G, Metri NJ, Opie RS, Jacka FN, Hawrelak JA, Rucklidge JJ, Ng CH, Sarris J. Effects of a high-prebiotic diet versus probiotic supplements versus synbiotics on adult mental health: The "Gut Feelings" randomised controlled trial. Front Neurosci 2023; 16:1097278. [PMID: 36815026 PMCID: PMC9940791 DOI: 10.3389/fnins.2022.1097278] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/15/2022] [Indexed: 02/09/2023] Open
Abstract
Background Preliminary evidence supports the use of dietary interventions and gut microbiota-targeted interventions such as probiotic or prebiotic supplementation for improving mental health. We report on the first randomised controlled trial (RCT) to examine the effects of a high-prebiotic dietary intervention and probiotic supplements on mental health. Methods "Gut Feelings" was an 8-week, 2 × 2 factorial RCT of 119 adults with moderate psychological distress and low prebiotic food intake. Treatment arms: (1) probiotic supplement and diet-as-usual (probiotic group); (2) high-prebiotic diet and placebo supplement (prebiotic diet group); (3) probiotic supplement and high-prebiotic diet (synbiotic group); and (4) placebo supplement and diet-as-usual (placebo group). The primary outcome was assessment of total mood disturbance (TMD; Profile of Mood States Short Form) from baseline to 8 weeks. Secondary outcomes included anxiety, depression, stress, sleep, and wellbeing measures. Results A modified intention-to-treat analysis using linear mixed effects models revealed that the prebiotic diet reduced TMD relative to placebo at 8 weeks [Cohen's d = -0.60, 95% confidence interval (CI) = -1.18, -0.03; p = 0.039]. There was no evidence of symptom improvement from the probiotic (d = -0.19, 95% CI = -0.75, 0.38; p = 0.51) or synbiotic treatments (d = -0.03, 95% CI = -0.59, 0.53; p = 0.92). Improved anxiety, stress, and sleep were noted in response to the prebiotic diet while the probiotic tentatively improved wellbeing, relative to placebo. No benefit was found in response to the synbiotic intervention. All treatments were well tolerated with few adverse events. Conclusion A high-prebiotic dietary intervention may improve mood, anxiety, stress, and sleep in adults with moderate psychological distress and low prebiotic intake. A synbiotic combination of high-prebiotic diet and probiotic supplement does not appear to have a beneficial effect on mental health outcomes, though further evidence is required. Results are limited by the relatively small sample size. Clinical trial registration https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372753, identifier ACTRN12617000795392.
Collapse
Affiliation(s)
- Tanya M. Freijy
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia,Faculty of Medicine, Dentistry and Health Sciences, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Lachlan Cribb
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Georgina Oliver
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Najwa-Joelle Metri
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Rachelle S. Opie
- IPAN, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, VIC, Australia
| | - Felice N. Jacka
- School of Medicine, Food and Mood Centre, IMPACT Strategic Research Centre, Deakin University, Melbourne, VIC, Australia,Centre for Adolescent Health, Murdoch Children’s Research Institute, Melbourne, VIC, Australia,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, OLD, Australia
| | - Jason A. Hawrelak
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia,Human Nutrition and Functional Medicine Department, University of Western States, Portland, OR, United States
| | - Julia J. Rucklidge
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Chee H. Ng
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Jerome Sarris
- Professorial Unit, The Melbourne Clinic, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia,Faculty of Medicine, Dentistry and Health Sciences, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia,NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia,*Correspondence: Jerome Sarris,
| |
Collapse
|
17
|
Saleh-Ghadimi S, Dehghan P, Sarmadi B, Maleki P. Improvement of sleep by resistant dextrin prebiotic in type 2 diabetic women coincides with attenuation of metabolic endotoxemia: involvement of gut-brain axis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:5229-5237. [PMID: 35306660 DOI: 10.1002/jsfa.11876] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 03/12/2022] [Accepted: 03/20/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Resistant dextrin, as a prebiotic and functional food, may possess favorable effects in type 2 diabetes. This study was conducted to assess whether supplementation with resistant dextrin can improve sleep and quality of life in obese type 2 diabetic women. RESULTS In this randomized controlled trial, female obese type 2 diabetic patients (n = 76) were randomly assigned into intervention group (n = 38) and placebo group (n = 38), and received 10 g day-1 of resistant dextrin or maltodextrin for a period of 8 weeks, respectively. Sleep quality and quality of life (QOL) were assessed by Pittsburgh Sleep Quality Index (PSQI) and SF-36 health survey, respectively. Fasting blood samples were driven to measure serum bacterial endotoxin, fasting blood sugar, glycosylated hemoglobin (HbA1c), pro-inflammatory/anti-inflammatory biomarkers (IL-18, IL-6, IL-10, TNF-α), and biomarkers of hypothalamic-pituitary-adrenal (HPA) axis function [tryptophan (TRP), adrenocorticotropic hormone (ACTH), kynurenine (KYN), cortisol]. Supplementation with resistant dextrin improved sleep (P < 0.001) and QOL (P < 0.001) significantly. It also caused a significant decrease in levels of endotoxin, HbA1c, IL-18, IL-6, TNF-α and a significant increase in IL-10 levels. Significant and positive correlations were found between endotoxin (r = 0.488, P = 0.003), IL-6 (r = 0.436, P = 0.008), IL-18 (r = 0.475, P = 0.003), cortisol (r = 0.545, P = 0.048), KYN/TRP (r = 0.527, P = 0.001), and PSQI scores. CONCLUSIONS It is concluded that resistant dextrin improves sleep and QOL in obese women with type 2 diabetes. Its beneficial effects may be attributed in part to modulation of glycemia, metabolic endotoxemia and subsequently a decrease in biomarkers of inflammation and HPA axis activity. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Sevda Saleh-Ghadimi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Dehghan
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Sarmadi
- Department of Nutrition Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Parham Maleki
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
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
|
19
|
Haarhuis JE, Kardinaal A, Kortman GAM. Probiotics, prebiotics and postbiotics for better sleep quality: a narrative review. Benef Microbes 2022; 13:169-182. [PMID: 35815493 DOI: 10.3920/bm2021.0122] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is a growing prevalence of sleep problems and insomnia worldwide, urging the development of new treatments to tackle this increase. Several studies have suggested that the gut microbiome might influence sleep quality. The gut microbiome affects the host's health via the production of metabolites and compounds with neuroactive and immunomodulatory properties, which include short-chain fatty acids, secondary bile acids and neurotransmitters. Several of these metabolites and compounds are independently known as wakefulness-promoting (serotonin, epinephrine, dopamine, orexin, histamine, acetylcholine, cortisol) or sleep-promoting (gamma-aminobutyric acid, melatonin). The primary aim of this review was to evaluate the potential of pro-, pre- and postbiotic treatments to improve sleep quality. Additionally, we aimed to evaluate whether each of the treatments could ameliorate stress and anxiety, which are known to bidirectionally correlate with sleep problems. Lastly, we provided a mechanistic explanation for our findings. A literature search was conducted using PubMed, Scopus, Web of Science, and Google Scholar to compare all human trials that met our inclusion criteria and were published before November 2021. We furthermore discussed relevant findings from animal experiments to provide a mechanistic insight. While several studies found that sleep latency, sleep length, and cortisol levels improved after pro-, pre- or postbiotic treatment, others did not show any significant improvements for sleep quality, stress, or anxiety. These discrepancies can be explained by between-study variations in study designs, study populations, treatments, type and level of distress, and sex differences. We conclude that the trials discussed provide some evidence for prebiotics, postbiotics, and traditional probiotics, such as those belonging to lactobacilli and bifidobacteria, to improve sleep quality and stress, but stronger evidence might be found in the future after implementing the methodological adjustments that are suggested in this review.
Collapse
Affiliation(s)
- J E Haarhuis
- Division of Human Nutrition and Health, Wageningen University, P.O. Box 17, 6700 AA Wageningen, the Netherlands.,Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
| | - A Kardinaal
- NIZO food research B.V., Kernhemseweg 2, 6718 ZB Ede, the Netherlands
| | - G A M Kortman
- NIZO food research B.V., Kernhemseweg 2, 6718 ZB Ede, the Netherlands
| |
Collapse
|
20
|
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
|
21
|
Bowers SJ, Summa KC, Thompson RS, González A, Vargas F, Olker C, Jiang P, Lowry CA, Dorrestein PC, Knight R, Wright KP, Fleshner M, Turek FW, Vitaterna MH. A Prebiotic Diet Alters the Fecal Microbiome and Improves Sleep in Response to Sleep Disruption in Rats. Front Neurosci 2022; 16:889211. [PMID: 35685770 PMCID: PMC9172596 DOI: 10.3389/fnins.2022.889211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/28/2022] [Indexed: 12/16/2022] Open
Abstract
Sleep disruption is a challenging and exceedingly common physiological state that contributes to a wide range of biochemical and molecular perturbations and has been linked to numerous adverse health outcomes. Modern society exerts significant pressure on the sleep/wake cycle via myriad factors, including exposure to electric light, psychological stressors, technological interconnection, jet travel, shift work, and widespread use of sleep-affecting compounds. Interestingly, recent research has identified a link between the microbiome and the regulation of sleep, suggesting that interventions targeting the microbiome may offer unique therapeutic approaches to challenges posed by sleep disruption. In this study, we test the hypothesis that administration of a prebiotic diet containing galactooligosaccharides (GOS) and polydextrose (PDX) in adult male rats improves sleep in response to repeated sleep disruption and during recovery sleep. We found that animals fed the GOS/PDX prebiotic diet for 4 weeks exhibit increased non-rapid eye movement (NREM) and rapid eye movement (REM) sleep during 5 days of sleep disruption and increased total sleep time during 24 h of recovery from sleep disruption compared to animals fed a control diet, despite similar baseline sleep characteristics. Further, the GOS/PDX prebiotic diet led to significant changes in the fecal microbiome. Consistent with previous reports, the prebiotic diet increased the relative abundance of the species Parabacteroides distasonis, which positively correlated with sleep parameters during recovery sleep. Taken together, these findings suggest that the GOS/PDX prebiotic diet may offer an approach to improve resilience to the physiologic challenge of sleep disruption, in part through impacts on the microbiome.
Collapse
Affiliation(s)
- Samuel J. Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University Weinberg College of Arts and Sciences, Evanston, IL, United States
| | - Keith C. Summa
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Division of Gastroenterology & Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Robert S. Thompson
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado, Boulder, Boulder, CO, United States
| | - Antonio González
- Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, CA, United States
| | - Fernando Vargas
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Christopher Olker
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University Weinberg College of Arts and Sciences, Evanston, IL, United States
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University Weinberg College of Arts and Sciences, Evanston, IL, United States
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado, Boulder, Boulder, CO, United States
| | - Pieter C. Dorrestein
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, United States
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, CA, United States
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, United States
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado, Boulder, Boulder, CO, United States
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, CO, United States
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, United States
- Center for Neuroscience, University of Colorado, Boulder, Boulder, CO, United States
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University Weinberg College of Arts and Sciences, Evanston, IL, United States
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Martha H. Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University Weinberg College of Arts and Sciences, Evanston, IL, United States
| |
Collapse
|
22
|
Thum C, Wall C, Day L, Szeto IMY, Li F, Yan Y, Barnett MPG. Changes in Human Milk Fat Globule Composition Throughout Lactation: A Review. Front Nutr 2022; 9:835856. [PMID: 35634409 PMCID: PMC9137899 DOI: 10.3389/fnut.2022.835856] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/22/2022] [Indexed: 01/19/2023] Open
Abstract
There has been a growing interest in understanding how the relative levels of human milk fat globule (MFG) components change over the course of lactation, how they differ between populations, and implications of these changes for the health of the infant. In this article, we describe studies published over the last 30 years which have investigated components of the MFG in term milk, focusing on changes over the course of lactation and highlighting infant and maternal factors that may influence these changes. We then consider how the potential health benefits of some of the milk fat globule membrane (MFGM) components and derived ingredients relate to compositional and functional aspects and how these change throughout lactation. The results show that the concentrations of phospholipids, gangliosides, cholesterol, fatty acids and proteins vary throughout lactation, and such changes are likely to reflect the changing requirements of the growing infant. There is a lack of consistent trends for changes in phospholipids and gangliosides across lactation which may reflect different methodological approaches. Other factors such as maternal diet and geographical location have been shown to influence human MFGM composition. The majority of research on the health benefits of MFGM have been conducted using MFGM ingredients derived from bovine milk, and using animal models which have clearly demonstrated the role of the MFGM in supporting cognitive and immune health of infants at different stages of growth and development.
Collapse
Affiliation(s)
- Caroline Thum
- AgResearch Ltd, Te Ohu Rangahau Kai, Palmerston North, New Zealand
- *Correspondence: Caroline Thum
| | - Clare Wall
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Li Day
- AgResearch Ltd, Te Ohu Rangahau Kai, Palmerston North, New Zealand
| | - Ignatius M. Y. Szeto
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group, Co., Ltd, Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co., Ltd, Hohhot, China
| | - Fang Li
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group, Co., Ltd, Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co., Ltd, Hohhot, China
| | - Yalu Yan
- Yili Maternal and Infant Nutrition Institute, Inner Mongolia Yili Industrial Group, Co., Ltd, Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co., Ltd, Hohhot, China
| | | |
Collapse
|
23
|
Ditmer M, Gabryelska A, Turkiewicz S, Białasiewicz P, Małecka-Wojciesko E, Sochal M. Sleep Problems in Chronic Inflammatory Diseases: Prevalence, Treatment, and New Perspectives: A Narrative Review. J Clin Med 2021; 11:67. [PMID: 35011807 PMCID: PMC8745687 DOI: 10.3390/jcm11010067] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/15/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Epidemiological studies have shown that individuals with sleep problems are at a greater risk of developing immune and chronic inflammatory diseases. As sleep disorders and low sleep quality in the general population are frequent ailments, it seems important to recognize them as serious public health problems. The exact relation between immunity and sleep remains elusive; however, it might be suspected that it is shaped by others stress and alterations of the circadian rhythm (commonly caused by for example shift work). As studies show, drugs used in the therapy of chronic inflammatory diseases, such as steroids or monoclonal antibodies, also influence sleep in more complex ways than those resulting from attenuation of the disease symptoms. Interestingly, the relation between sleep and immunity appears to be bidirectional; that is, sleep may influence the course of immune diseases, such as inflammatory bowel disease. Thus, proper diagnosis and treatment of sleep disorders are vital to the patient's immune status and, in effect, health. This review examines the epidemiology of sleep disorders and immune diseases, the associations between them, and their current treatment and novel perspectives in therapy.
Collapse
Affiliation(s)
- Marta Ditmer
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (M.D.); (A.G.); (S.T.); (P.B.)
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (M.D.); (A.G.); (S.T.); (P.B.)
| | - Szymon Turkiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (M.D.); (A.G.); (S.T.); (P.B.)
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (M.D.); (A.G.); (S.T.); (P.B.)
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (M.D.); (A.G.); (S.T.); (P.B.)
| |
Collapse
|
24
|
Thompson RS, Gaffney M, Hopkins S, Kelley T, Gonzalez A, Bowers SJ, Vitaterna MH, Turek FW, Foxx CL, Lowry CA, Vargas F, Dorrestein PC, Wright KP, Knight R, Fleshner M. Ruminiclostridium 5, Parabacteroides distasonis, and bile acid profile are modulated by prebiotic diet and associate with facilitated sleep/clock realignment after chronic disruption of rhythms. Brain Behav Immun 2021; 97:150-166. [PMID: 34242738 DOI: 10.1016/j.bbi.2021.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic disruption of rhythms (CDR) impacts sleep and can result in circadian misalignment of physiological systems which, in turn, is associated with increased disease risk. Exposure to repeated or severe stressors also disturbs sleep and diurnal rhythms. Prebiotic nutrients produce favorable changes in gut microbial ecology, the gut metabolome, and reduce several negative impacts of acute severe stressor exposure, including disturbed sleep, core body temperature rhythmicity, and gut microbial dysbiosis. In light of previous compelling evidence that prebiotic diet broadly reduces negative impacts of acute, severe stressors, we hypothesize that prebiotic diet will also effectively mitigate the negative impacts of chronic disruption of circadian rhythms on physiology and sleep/wake behavior. Male, Sprague Dawley rats were fed diets enriched in prebiotic substrates or calorically matched control chow. After 5 weeks on diet, rats were exposed to CDR (12 h light/dark reversal, weekly for 8 weeks) or remained on undisturbed normal light/dark cycles (NLD). Sleep EEG, core body temperature, and locomotor activity were recorded via biotelemetry in freely moving rats. Fecal samples were collected on experimental days -33, 0 (day of onset of CDR), and 42. Taxonomic identification and relative abundances of gut microbes were measured in fecal samples using 16S rRNA gene sequencing and shotgun metagenomics. Fecal primary, bacterially modified secondary, and conjugated bile acids were measured using liquid chromatography with tandem mass spectrometry (LC-MS/MS). Prebiotic diet produced rapid and stable increases in the relative abundances of Parabacteroides distasonis and Ruminiclostridium 5. Shotgun metagenomics analyses confirmed reliable increases in relative abundances of Parabacteroides distasonis and Clostridium leptum, a member of the Ruminiclostridium genus. Prebiotic diet also modified fecal bile acid profiles; and based on correlational and step-wise regression analyses, Parabacteroides distasonis and Ruminiclostridium 5 were positively associated with each other and negatively associated with secondary and conjugated bile acids. Prebiotic diet, but not CDR, impacted beta diversity. Measures of alpha diversity evenness were decreased by CDR and prebiotic diet prevented that effect. Rats exposed to CDR while eating prebiotic, compared to control diet, more quickly realigned NREM sleep and core body temperature (ClockLab) diurnal rhythms to the altered light/dark cycle. Finally, both cholic acid and Ruminiclostridium 5 prior to CDR were associated with time to realign CBT rhythms to the new light/dark cycle after CDR; whereas both Ruminiclostridium 5 and taurocholic acid prior to CDR were associated with NREM sleep recovery after CDR. These results support our hypothesis and suggest that ingestion of prebiotic substrates is an effective strategy to increase the relative abundance of health promoting microbes, alter the fecal bile acid profile, and facilitate the recovery and realignment of sleep and diurnal rhythms after circadian disruption.
Collapse
Affiliation(s)
- Robert S Thompson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA; Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| | - Michelle Gaffney
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA; Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Shelby Hopkins
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Tel Kelley
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Samuel J Bowers
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL, USA
| | - Martha Hotz Vitaterna
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL, USA
| | - Fred W Turek
- Department of Neurobiology, Northwestern University, Center for Sleep and Circadian Biology, Evanston, IL, USA
| | - Christine L Foxx
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA; Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Fernando Vargas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA
| | - Kenneth P Wright
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA; Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA; Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| |
Collapse
|
25
|
Pacheco AP, Cedernaes J, Benedict C. Gut microbiome as a therapeutic target in the treatment of sleep disorders: where we are. Sleep Med Rev 2021; 60:101547. [PMID: 34571476 DOI: 10.1016/j.smrv.2021.101547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 11/30/2022]
Affiliation(s)
- André P Pacheco
- Department of Neuroscience (Sleep Science, BMC), Uppsala University, Uppsala, Sweden
| | | | - Christian Benedict
- Department of Neuroscience (Sleep Science, BMC), Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Clouard C, Reimert I, Fleming SA, Koopmans SJ, Schuurman T, Hauser J. Dietary sialylated oligosaccharides in early-life may promote cognitive flexibility during development in context of obesogenic dietary intake. Nutr Neurosci 2021; 25:2461-2478. [PMID: 34565309 DOI: 10.1080/1028415x.2021.1975877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Oligosaccharides found in mammalian milk have shown the potential to alter brain development across multiple species. The diversity and concentration of these oligosaccharides is species-specific and varies greatly between individuals, thus understanding their role in cognitive development is warranted. We investigated the impact of early life dietary fucosylated/neutral or sialylated human milk oligosaccharides (HMO) on behaviours in tasks assessing anxiety, motivation, appetite, learning, and memory.Methods: Sixty-four female Göttingen minipigs were artificially reared from 2 weeks postnatal and provided milk replacers. The study used four groups: no additional oligosaccharides (Con), fucosylated and neutral oligosaccharides (FN, 4 g/L), sialylated oligosaccharides (SL, 0.68 g/L), or both FN and SL (FN + SL, 4 g/L) from 2 to 11 weeks postnatal. One reference group was sow-reared. Weaning occurred between 10 and 11 weeks postnatal, and thereafter an obesogenic diet was provided. Behavioral tasks were conducted over three periods: 1) 0-11 weeks; 2) 16-29 weeks; 3) 39-45 weeks. Tasks included a spatial holeboard task, open field task, exposure to a novel object, runway task, single-feed task, and home pen behaviour observation.Results: In the holeboard, the SL group demonstrated improved reference memory during reversal trials between 16-29 weeks. All groups demonstrated equivalent behavior in open field, novel object, runway, and single-feed tasks, as well as in their home pens (Ps > 0.05).Discussion: These results suggest that early life dietary intake of sialylated oligosaccharides may provide an improvement to cognition during the equivalent developmental stage of adolescence.
Collapse
Affiliation(s)
- Caroline Clouard
- Department of Animal Sciences, Adaptation Physiology Group, Wageningen University & Research, Wageningen, Netherlands.,PEGASE, INRAE, Institut Agro, Saint-Gilles, France
| | - Inonge Reimert
- Department of Animal Sciences, Adaptation Physiology Group, Wageningen University & Research, Wageningen, Netherlands
| | | | - Sietse-Jan Koopmans
- Wageningen Livestock Research, Wageningen University & Research, Netherlands
| | - Teun Schuurman
- Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Jonas Hauser
- Brain Health, Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1005 Lausanne, Switzerland
| |
Collapse
|
27
|
Lin A, Shih CT, Chu HF, Chen CW, Cheng YT, Wu CC, Yang CCH, Tsai YC. Lactobacillus fermentum PS150 promotes non-rapid eye movement sleep in the first night effect of mice. Sci Rep 2021; 11:16313. [PMID: 34381098 PMCID: PMC8357945 DOI: 10.1038/s41598-021-95659-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/06/2021] [Indexed: 12/30/2022] Open
Abstract
The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3–V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.
Collapse
Affiliation(s)
- Alexander Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Chung Mei Biopharma Co., Ltd., Taichung, Taiwan
| | | | - Hsu-Feng Chu
- Biomedical Industry Ph.D. Program, National Yang-Ming University, Taipei, Taiwan
| | - Chieh-Wen Chen
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Cheng
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan
| | | | - Cheryl C H Yang
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan. .,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan. .,Brain Research Center, National Yang-Ming University, Taipei, Taiwan.
| | - Ying-Chieh Tsai
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.
| |
Collapse
|
28
|
Sen P, Molinero-Perez A, O'Riordan KJ, McCafferty CP, O'Halloran KD, Cryan JF. Microbiota and sleep: awakening the gut feeling. Trends Mol Med 2021; 27:935-945. [PMID: 34364787 DOI: 10.1016/j.molmed.2021.07.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Various lifestyle and environmental factors are known to influence sleep. Increasingly, evidence points to a role for the microbiota in regulating brain and behaviour. This article explores how the microbiota-gut-brain axis affects sleep directly and indirectly. We summarize the possible molecular mechanisms underlying sleep-microbiome interactions and discuss how various factors interact with the gut microbiota to influence sleep. Furthermore, we present the current evidence of alterations of the microbiota-gut-brain axis in various sleep disorders and pathologies where comorbid sleep disturbances are common. Since manipulating the gut microbiota could potentially improve sleep, we outline ways in which this can be achieved.
Collapse
Affiliation(s)
- Paromita Sen
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | - Cian P McCafferty
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Physiology, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
29
|
Bastiaanssen TFS, Cussotto S, Claesson MJ, Clarke G, Dinan TG, Cryan JF. Gutted! Unraveling the Role of the Microbiome in Major Depressive Disorder. Harv Rev Psychiatry 2021; 28:26-39. [PMID: 31913980 PMCID: PMC7012351 DOI: 10.1097/hrp.0000000000000243] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microorganisms can be found in virtually any environment. In humans, the largest collection of microorganisms is found in the gut ecosystem. The adult gut microbiome consists of more genes than its human host and typically spans more than 60 genera from across the taxonomic tree. In addition, the gut contains the largest number of neurons in the body, after the brain. In recent years, it has become clear that the gut microbiome is in communication with the brain, through the gut-brain axis. A growing body of literature shows that the gut microbiome plays a shaping role in a variety of psychiatric disorders, including major depressive disorder (MDD). In this review, the interplay between the microbiome and MDD is discussed in three facets. First, we discuss factors that affect the onset/development of MDD that also greatly impinge on the composition of the gut microbiota-especially diet and stressful life events. We then examine the interplay between the microbiota and MDD. We examine evidence suggesting that the microbiota is altered in MDD, and we discuss why the microbiota should be considered during MDD treatment. Finally, we look toward the future and examine how the microbiota might become a therapeutic target for MDD. This review is intended to introduce those familiar with the neurological and psychiatric aspects of MDD to the microbiome and its potential role in the disorder. Although research is in its very early days, with much yet to be the understood, the microbiome is offering new avenues for developing potentially novel strategies for managing MDD.
Collapse
|
30
|
Chichlowski M, Bokulich N, Harris CL, Wampler JL, Li F, Berseth CL, Rudolph C, Wu SS. Effect of Bovine Milk Fat Globule Membrane and Lactoferrin in Infant Formula on Gut Microbiome and Metabolome at 4 Months of Age. Curr Dev Nutr 2021; 5:nzab027. [PMID: 33981943 PMCID: PMC8105244 DOI: 10.1093/cdn/nzab027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Milk fat globule membrane (MFGM) and lactoferrin (LF) are human-milk bioactive components demonstrated to support gastrointestinal and immune development. Significantly fewer diarrhea and respiratory-associated adverse events through 18 mo of age were previously reported in healthy term infants fed a cow-milk-based infant formula with an added source of bovine MFGM and bovine LF through 12 mo of age. OBJECTIVES The aim was to compare microbiota and metabolite profiles in a subset of study participants. METHODS Stool samples were collected at baseline (10-14 d of age) and day 120. Bacterial community profiling was performed via 16S rRNA gene sequencing and alpha and beta diversity were analyzed (QIIME 2). Differentially abundant taxa were determined using linear discriminant analysis effect size (LefSE) and visualized (Metacoder). Untargeted stool metabolites were analyzed (HPLC/MS) and expressed as the fold-change between group means (control to MFGM+LF ratio). RESULTS Alpha diversity increased significantly in both groups from baseline to 4 mo. Subtle group differences in beta diversity were demonstrated at 4 mo (Jaccard distance; R 2 = 0.01, P = 0.042). Specifically, Bacteroides uniformis and Bacteroides plebeius were more abundant in the MFGM+LF group at 4 mo. Metabolite profile differences for MFGM+LF versus control included lower fecal medium-chain fatty acids, deoxycarnitine, and glycochenodeoxycholate, and some higher fecal carbohydrates and steroids (P < 0.05). After applying multiple test correction, the differences in stool metabolomics were not significant. CONCLUSIONS Addition of bovine MFGM and LF in infant formula was associated with subtle differences in stool microbiome and metabolome by 4 mo of age, including increased prevalence of Bacteroides species. Stool metabolite profiles may be consistent with altered microbial metabolism. This trial was registered at https://clinicaltrials.gov as NCT02274883.
Collapse
Affiliation(s)
- Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
| | - Nicholas Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition, and Health, ETH Zürich, Zurich, Switzerland
| | - Cheryl L Harris
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
| | - Jennifer L Wampler
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
| | - Fei Li
- Departments of Developmental and Behavioral Pediatrics & Child Primary Care, MOE-Shanghai Key Lab for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Carol Lynn Berseth
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
| | - Colin Rudolph
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Steven S Wu
- Medical and Scientific Affairs, Reckitt|Mead Johnson Nutrition Institute, Evansville, IN, USA
- Division of Pediatric Gastroenterology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
31
|
Withrow D, Bowers SJ, Depner CM, González A, Reynolds AC, Wright KP. Sleep and Circadian Disruption and the Gut Microbiome-Possible Links to Dysregulated Metabolism. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2021; 17:26-37. [PMID: 34805616 PMCID: PMC8597978 DOI: 10.1016/j.coemr.2020.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Insufficient sleep and circadian misalignment are associated with adverse metabolic health outcomes. Alterations in gut microbial diversity occur with insufficient sleep and circadian misalignment, which can lead to modifications in microbial structure and function. Changes in microbially produced and modified metabolites such as short chain fatty acids and secondary bile acids may contribute to chronic inflammation, positive energy balance and endocrine changes, and represent potential mechanisms linking insufficient sleep and circadian misalignment with metabolic dysregulation. Literature primarily from the last two years is reviewed here, examining the impact of sleep and circadian rhythms and their disruption on the gut microbiome in human and non-human models, with an emphasis on the hypothesis that the altered gut microbiome may be one pathway by which insufficient sleep and circadian misalignment dysregulate metabolism.
Collapse
Affiliation(s)
- Dana Withrow
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO, USA
| | - Samuel J. Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, USA
| | - Christopher M. Depner
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO, USA
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, USA
| | - Antonio González
- Department of Pediatrics, University of California at San Diego, La Jolla, CA, USA
| | - Amy C. Reynolds
- The Appleton Institute, CQUniversity Australia, Adelaide, Australia
| | - Kenneth P. Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO, USA
| |
Collapse
|
32
|
Yeruva L, Munblit D, Collado MC. Editorial: Impact of Early Life Nutrition on Immune System Development and Related Health Outcomes in Later Life. Front Immunol 2021; 12:668569. [PMID: 33841449 PMCID: PMC8027300 DOI: 10.3389/fimmu.2021.668569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
- Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Daniel Munblit
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom.,Solov'ev Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - Maria Carmen Collado
- Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
33
|
Brown T, Sykes D, Allen AR. Implications of Breast Cancer Chemotherapy-Induced Inflammation on the Gut, Liver, and Central Nervous System. Biomedicines 2021; 9:biomedicines9020189. [PMID: 33668580 PMCID: PMC7917715 DOI: 10.3390/biomedicines9020189] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Breast Cancer is still one of the most common cancers today; however, with advancements in diagnostic and treatment methods, the mortality and survivorship of patients continues to decrease and increase, respectively. Commonly used treatments today consist of drug combinations, such as doxorubicin and cyclophosphamide; docetaxel, doxorubicin, and cyclophosphamide; or doxorubicin, cyclophosphamide, and paclitaxel. Although these combinations are effective at destroying cancer cells, there is still much to be understood about the effects that chemotherapy can have on normal organ systems such as the nervous system, gastrointestinal tract, and the liver. Patients can experience symptoms of cognitive impairments or “chemobrain”, such as difficulty in concentrating, memory recollection, and processing speed. They may also experience gastrointestinal (GI) distress symptoms such as diarrhea and vomiting, as well as hepatotoxicity and long term liver damage. Chemotherapy treatment has also been shown to induce peripheral neuropathy resulting in numbing, pain, and tingling sensations in the extremities of patients. Interestingly, researchers have discovered that this array of symptoms that cancer patients experience are interconnected and mediated by the inflammatory response.
Collapse
Affiliation(s)
- Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - DeLawrence Sykes
- Department of Biology, Pomona College, Claremont, CA 91711, USA;
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: ; Tel.: +1-501-686-7335
| |
Collapse
|
34
|
Partrick KA, Rosenhauer AM, Auger J, Arnold AR, Ronczkowski NM, Jackson LM, Lord MN, Abdulla SM, Chassaing B, Huhman KL. Ingestion of probiotic (Lactobacillus helveticus and Bifidobacterium longum) alters intestinal microbial structure and behavioral expression following social defeat stress. Sci Rep 2021; 11:3763. [PMID: 33580118 PMCID: PMC7881201 DOI: 10.1038/s41598-021-83284-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
Social stress exacerbates anxious and depressive behaviors in humans. Similarly, anxiety- and depressive-like behaviors are triggered by social stress in a variety of non-human animals. Here, we tested whether oral administration of the putative anxiolytic probiotic strains Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces the striking increase in anxiety-like behavior and changes in gut microbiota observed following social defeat stress in Syrian hamsters. We administered the probiotic at two different doses for 21 days, and 16S rRNA gene amplicon sequencing revealed a shift in microbial structure following probiotic administration at both doses, independently of stress. Probiotic administration at either dose increased anti-inflammatory cytokines IL-4, IL-5, and IL-10 compared to placebo. Surprisingly, probiotic administration at the low dose, equivalent to the one used in humans, significantly increased social avoidance and decreased social interaction. This behavioral change was associated with a reduction in microbial richness in this group. Together, these results demonstrate that probiotic administration alters gut microbial composition and may promote an anti-inflammatory profile but that these changes may not promote reductions in behavioral responses to social stress.
Collapse
Affiliation(s)
- Katherine A Partrick
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Anna M Rosenhauer
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Jérémie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, QC, Canada
| | - Amanda R Arnold
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Nicole M Ronczkowski
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Lanaya M Jackson
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Magen N Lord
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Sara M Abdulla
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Benoit Chassaing
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA.,INSERM U1016, Team "Mucosal Microbiota in Chronic Inflammatory Diseases", CNRS UMR 8104, Université de Paris, Paris, France.,Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Kim L Huhman
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA.
| |
Collapse
|
35
|
Kosmerl E, Rocha-Mendoza D, Ortega-Anaya J, Jiménez-Flores R, García-Cano I. Improving Human Health with Milk Fat Globule Membrane, Lactic Acid Bacteria, and Bifidobacteria. Microorganisms 2021; 9:341. [PMID: 33572211 PMCID: PMC7914750 DOI: 10.3390/microorganisms9020341] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
The milk fat globule membrane (MFGM), the component that surrounds fat globules in milk, and its constituents have gained significant attention for their gut function, immune-boosting properties, and cognitive-development roles. The MFGM can directly interact with probiotic bacteria, such as bifidobacteria and lactic acid bacteria (LAB), through interactions with bacterial surface proteins. With these interactions in mind, increasing evidence supports a synergistic effect between MFGM and probiotics to benefit human health at all ages. This important synergy affects the survival and adhesion of probiotic bacteria through gastrointestinal transit, mucosal immunity, and neurocognitive behavior in developing infants. In this review, we highlight the current understanding of the co-supplementation of MFGM and probiotics with a specific emphasis on their interactions and colocalization in dairy foods, supporting in vivo and clinical evidence, and current and future potential applications.
Collapse
Affiliation(s)
| | | | | | - Rafael Jiménez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA; (E.K.); (D.R.-M.); (J.O.-A.)
| | - Israel García-Cano
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA; (E.K.); (D.R.-M.); (J.O.-A.)
| |
Collapse
|
36
|
Colombo J, Carlson SE, Algarín C, Reyes S, Chichlowski M, Harris CL, Wampler JL, Peirano P, Berseth CL. Developmental effects on sleep-wake patterns in infants receiving a cow's milk-based infant formula with an added prebiotic blend: a Randomized Controlled Trial. Pediatr Res 2021; 89:1222-1231. [PMID: 32615579 PMCID: PMC8119237 DOI: 10.1038/s41390-020-1044-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/10/2020] [Accepted: 06/03/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Few studies have evaluated nutritive effects of prebiotics on infant behavior state, physiology, or metabolic status. METHODS In this double-blind randomized study, infants (n = 161) received cow's milk-based infant formula (Control) or similar formula with an added prebiotic blend (polydextrose and galactooligosaccharides [PDX/GOS]) from 14-35 to 112 days of age. Infant wake behavior (crying/fussing, awake/content) and 24-h sleep-wake actograms were analyzed (Baseline, Days 70 and 112). Salivary cortisol was immunoassayed (Days 70 and 112). In a subset, exploratory stool 16S ribosomal RNA-sequencing was analyzed (Baseline, Day 112). RESULTS One hundred and thirty-one infants completed the study. Average duration of crying/fussing episodes was similar at Baseline, significantly shorter for PDX/GOS vs. Control at Day 70, and the trajectory continued at Day 112. Latency to first and second nap was significantly longer for PDX/GOS vs. Control at Day 112. Cortisol awakening response was demonstrated at Days 70 and 112. Significant stool microbiome beta-diversity and individual taxa abundance differences were observed in the PDX/GOS group. CONCLUSIONS Results indicate faster consolidation of daytime waking state in infants receiving prebiotics and support home-based actigraphy to assess early sleep-wake patterns. A prebiotic effect on wake organization is consistent with influence on the gut-brain axis and warrants further investigation. IMPACT Few studies have evaluated nutritive effects of prebiotics on infant behavior state, cortisol awakening response, sleep-wake entrainment, and gut microbiome. Faster consolidation of daytime waking state was demonstrated in infants receiving a prebiotic blend in infant formula through ~4 months of age. Shorter episodes of crying were demonstrated at ~2 months of age (time point corresponding to age/developmental range associated with peak crying) in infants receiving formula with added prebiotics. Results support home-based actigraphy as a suitable method to assess early sleep-wake patterns. Prebiotic effect on wake organization is consistent with influence on the gut-brain axis and warrants further investigation.
Collapse
Affiliation(s)
- John Colombo
- Schiefelbusch Institute for Life Span Studies and Department of Psychology, University of Kansas, Lawrence, KS, USA.
| | - Susan E. Carlson
- grid.412016.00000 0001 2177 6375Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS USA
| | - Cecilia Algarín
- grid.443909.30000 0004 0385 4466Sleep and Functional Neurobiology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Sussanne Reyes
- grid.443909.30000 0004 0385 4466Sleep and Functional Neurobiology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Maciej Chichlowski
- Nutrition Science, Department of Medical Affairs, Mead Johnson Nutrition, 2400 West Lloyd Expy, Evansville, IN 47721 USA
| | - Cheryl L. Harris
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, 2400 West Lloyd Expy, Evansville, IN 47721 USA
| | - Jennifer L. Wampler
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, 2400 West Lloyd Expy, Evansville, IN 47721 USA
| | - Patricio Peirano
- grid.443909.30000 0004 0385 4466Sleep and Functional Neurobiology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Carol Lynn Berseth
- Clinical Research, Department of Medical Affairs, Mead Johnson Nutrition, 2400 West Lloyd Expy, Evansville, IN 47721 USA
| |
Collapse
|
37
|
Gut microbiota depletion by chronic antibiotic treatment alters the sleep/wake architecture and sleep EEG power spectra in mice. Sci Rep 2020; 10:19554. [PMID: 33177599 PMCID: PMC7659342 DOI: 10.1038/s41598-020-76562-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
Dysbiosis of the gut microbiota affects physiological processes, including brain functions, by altering the intestinal metabolism. Here we examined the effects of the gut microbiota on sleep/wake regulation. C57BL/6 male mice were treated with broad-spectrum antibiotics for 4 weeks to deplete their gut microbiota. Metabolome profiling of cecal contents in antibiotic-induced microbiota-depleted (AIMD) and control mice showed significant variations in the metabolism of amino acids and vitamins related to neurotransmission, including depletion of serotonin and vitamin B6, in the AIMD mice. Sleep analysis based on electroencephalogram and electromyogram recordings revealed that AIMD mice spent significantly less time in non-rapid eye movement sleep (NREMS) during the light phase while spending more time in NREMS and rapid eye movement sleep (REMS) during the dark phase. The number of REMS episodes seen in AIMD mice increased during both light and dark phases, and this was accompanied by frequent transitions from NREMS to REMS. In addition, the theta power density during REMS was lower in AIMD mice during the light phase compared with that in the controls. Consequently, the gut microbiota is suggested to affect the sleep/wake architecture by altering the intestinal balance of neurotransmitters.
Collapse
|
38
|
Abd El‐Salam MH, El‐Shibiny S. Milk fat globule membrane: An overview with particular emphasis on its nutritional and health benefits. INT J DAIRY TECHNOL 2020. [DOI: 10.1111/1471-0307.12730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Safinaz El‐Shibiny
- Dairy Department National Research Centre El‐Behous St Dokki Cairo Egypt
| |
Collapse
|
39
|
O'Connor KM, Lucking EF, Bastiaanssen TFS, Peterson VL, Crispie F, Cotter PD, Clarke G, Cryan JF, O'Halloran KD. Prebiotic administration modulates gut microbiota and faecal short-chain fatty acid concentrations but does not prevent chronic intermittent hypoxia-induced apnoea and hypertension in adult rats. EBioMedicine 2020; 59:102968. [PMID: 32861200 PMCID: PMC7475129 DOI: 10.1016/j.ebiom.2020.102968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/22/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Evidence is accruing to suggest that microbiota-gut-brain signalling plays a regulatory role in cardiorespiratory physiology. Chronic intermittent hypoxia (CIH), modelling human sleep apnoea, affects gut microbiota composition and elicits cardiorespiratory morbidity. We investigated if treatment with prebiotics ameliorates cardiorespiratory dysfunction in CIH-exposed rats. METHODS Adult male rats were exposed to CIH (96 cycles/day, 6.0% O2 at nadir) for 14 consecutive days with and without prebiotic supplementation (fructo- and galacto-oligosaccharides) beginning two weeks prior to gas exposures. FINDINGS CIH increased apnoea index and caused hypertension. CIH exposure had modest effects on the gut microbiota, decreasing the relative abundance of Lactobacilli species, but had no effect on microbial functional characteristics. Faecal short-chain fatty acid (SCFA) concentrations, plasma and brainstem pro-inflammatory cytokine concentrations and brainstem neurochemistry were unaffected by exposure to CIH. Prebiotic administration modulated gut microbiota composition and diversity, altering gut-metabolic (GMMs) and gut-brain (GBMs) modules and increased faecal acetic and propionic acid concentrations, but did not prevent adverse CIH-induced cardiorespiratory phenotypes. INTERPRETATION CIH-induced cardiorespiratory dysfunction is not dependant upon changes in microbial functional characteristics and decreased faecal SCFA concentrations. Prebiotic-related modulation of microbial function and resultant increases in faecal SCFAs were not sufficient to prevent CIH-induced apnoea and hypertension in our model. Our results do not exclude the potential for microbiota-gut-brain axis involvement in OSA-related cardiorespiratory morbidity, but they demonstrate that in a relatively mild model of CIH, sufficient to evoke classic cardiorespiratory dysfunction, such changes are not obligatory for the development of morbidity, but may become relevant in the elaboration and maintenance of cardiorespiratory morbidity with progressive disease. FUNDING Department of Physiology and APC Microbiome Ireland, University College Cork, Ireland. APC Microbiome Ireland is funded by Science Foundation Ireland, through the Government's National Development Plan.
Collapse
Affiliation(s)
- Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Fiona Crispie
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
40
|
Triplett J, Ellis D, Braddock A, Roberts E, Ingram K, Perez E, Short A, Brown D, Hutzley V, Webb C, Soto A, Chan V. Temporal and region-specific effects of sleep fragmentation on gut microbiota and intestinal morphology in Sprague Dawley rats. Gut Microbes 2020; 11:706-720. [PMID: 31924109 PMCID: PMC7524289 DOI: 10.1080/19490976.2019.1701352] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Sleep is a fundamental biological process, that when repeatedly disrupted, can result in severe health consequences. Recent studies suggest that both sleep fragmentation (SF) and dysbiosis of the gut microbiome can lead to metabolic disorders, though the underlying mechanisms are largely unclear. To better understand the consequences of SF, we investigated the effects of acute (6 days) and chronic (6 weeks) SF on rats by examining taxonomic profiles of microbiota in the distal ileum, cecum and proximal colon, as well as assessing structural and functional integrity of the gastrointestinal barrier. We further assayed the impact of SF on a host function by evaluating inflammation and immune response. Both acute and chronic SF induced microbial dysbiosis, more dramatically in the distal ileum (compared to other two regions studied), as noted by significant perturbations in alpha- and beta-diversity; though, specific microbial populations were significantly altered throughout each of the three regions. Furthermore, chronic SF resulted in increased crypt depth in the distal ileum and an increase in the number of villi lining both the cecum and proximal colon. Additional changes were noted with chronic SF, including: decreased microbial adhesion and penetration in the distal ileum and cecum, elevation in serum levels of the cytokine KC/GRO, and depressed levels of corticotropin. Importantly, our data show that perturbations to microbial ecology and intestinal morphology intensify in response to prolonged SF and these changes are habitat specific. Together, these results reveal consequences to gut microbiota homeostasis and host response following acute and chronic SF in rats.
Collapse
Affiliation(s)
- Judy Triplett
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - David Ellis
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amber Braddock
- Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Wright-Patterson AFB, OH, USA
| | - Erin Roberts
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Katherine Ingram
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Eric Perez
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amanda Short
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Dominique Brown
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victoria Hutzley
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Chelsey Webb
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Armando Soto
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victor Chan
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA,CONTACT Victor Chan Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| |
Collapse
|
41
|
Fontecha J, Brink L, Wu S, Pouliot Y, Visioli F, Jiménez-Flores R. Sources, Production, and Clinical Treatments of Milk Fat Globule Membrane for Infant Nutrition and Well-Being. Nutrients 2020; 12:E1607. [PMID: 32486129 PMCID: PMC7352329 DOI: 10.3390/nu12061607] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022] Open
Abstract
Research on milk fat globule membrane (MFGM) is gaining traction. The interest is two-fold; on the one hand, it is a unique trilayer structure with specific secretory function. On the other hand, it is the basis for ingredients with the presence of phospho- and sphingolipids and glycoproteins, which are being used as food ingredients with valuable functionality, in particular, for use as a supplement in infant nutrition. This last application is at the center of this Review, which aims to contribute to understanding MFGM's function in the proper development of immunity, cognition, and intestinal trophism, in addition to other potential effects such as prevention of diseases including cardiovascular disease, impaired bone turnover and inflammation, skin conditions, and infections as well as age-associated cognitive decline and muscle loss. The phospholipid composition of MFGM from bovine milk is quite like human milk and, although there are some differences due to dairy processing, these do not result in a chemical change. The MFGM ingredients, as used to improve the formulation in different clinical studies, have indeed increased the presence of phospholipids, sphingolipids, glycolipids, and glycoproteins with the resulting benefits of different outcomes (especially immune and cognitive outcomes) with no reported adverse effects. Nevertheless, the precise mechanism(s) of action of MFGM remain to be elucidated and further basic investigation is warranted.
Collapse
Affiliation(s)
- Javier Fontecha
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), 28049 Madrid, Spain
| | - Lauren Brink
- Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN 47721, USA; (L.B.); (S.W.)
| | - Steven Wu
- Department of Medical Affairs, Mead Johnson Nutrition, Evansville, IN 47721, USA; (L.B.); (S.W.)
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods (INAF), Department of Food Sciences, Laval University, Québec, QC G1V 0A6, Canada;
| | - Francesco Visioli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
- IMDEA-Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Rafael Jiménez-Flores
- Food Science and Technology Department, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
42
|
Schverer M, O'Mahony SM, O’Riordan KJ, Donoso F, Roy BL, Stanton C, Dinan TG, Schellekens H, Cryan JF. Dietary phospholipids: Role in cognitive processes across the lifespan. Neurosci Biobehav Rev 2020; 111:183-193. [DOI: 10.1016/j.neubiorev.2020.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/16/2019] [Accepted: 01/10/2020] [Indexed: 12/31/2022]
|
43
|
Thompson RS, Vargas F, Dorrestein PC, Chichlowski M, Berg BM, Fleshner M. Dietary prebiotics alter novel microbial dependent fecal metabolites that improve sleep. Sci Rep 2020; 10:3848. [PMID: 32123201 PMCID: PMC7051969 DOI: 10.1038/s41598-020-60679-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/13/2020] [Indexed: 01/01/2023] Open
Abstract
Dietary prebiotics produce favorable changes in the commensal gut microbiome and reduce host vulnerability to stress-induced disruptions in complex behaviors such as sleep. The mechanisms for how prebiotics modulate stress physiology remain unclear; however, emerging evidence suggests that gut microbes and their metabolites may play a role. This study tested if stress and/or dietary prebiotics (Test diet) alter the fecal metabolome; and explored if these changes were related to sleep and/or gut microbial alpha diversity. Male F344 rats on either Test or Control diet were instrumented for electroencephalography biotelemetry measures of sleep/wake. After 5 weeks on diet, rats were either stressed or remained in home cages. Based on untargeted mass spectrometry and 16S rRNA gene sequencing, both stress and Test diet altered the fecal metabolome/microbiome. In addition, Test diet prevented the stress-induced reduction in microbial alpha diversity based on PD_Whole_Tree, which has been previously published. Network propagation analysis revealed that stress increased members of the neuroactive steroidal pregnane molecular family; and that Test diet reduced this effect. We also discovered links between sleep, alpha diversity, and pyrimidine, secondary bile acid, and neuroactive glucocorticoid/pregnanolone-type steroidal metabolites. These results reveal novel microbial-dependent metabolites that may modulate stress physiology and sleep.
Collapse
Affiliation(s)
- Robert S Thompson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, 80309-0354, USA
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309-0354, USA
| | - Fernando Vargas
- Division of Biological Sciences, University of California, San Diego, CA, 92093, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, 92093, USA
| | - Pieter C Dorrestein
- Division of Biological Sciences, University of California, San Diego, CA, 92093, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, 92093, USA
| | | | - Brian M Berg
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN, 47712, USA
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, 80309-0354, USA.
- Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309-0354, USA.
| |
Collapse
|
44
|
Song D, Yang CS, Zhang X, Wang Y. The relationship between host circadian rhythms and intestinal microbiota: A new cue to improve health by tea polyphenols. Crit Rev Food Sci Nutr 2020; 61:139-148. [PMID: 31997655 DOI: 10.1080/10408398.2020.1719473] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Under the control of the host circadian rhythms, intestinal microbiota undergoes dietary-dependent diurnal fluctuations in composition and function. In addition, microbiome plays a critical role in maintaining the host circadian rhythms and metabolic homeostasis. The interactions between host circadian rhythms and intestinal microbiota suggest that intervention with prebiotics or probiotic is a possible way to alleviate circadian rhythm misalignment and related metabolic diseases. This review discusses the circadian rhythm oscillations of gut flora, relationship between host circadian rhythms and microbiome and related effects on metabolism. The influence on circadian rhythms by the interactions between tea polyphenols (TP) and intestinal microbiota is highlighted.
Collapse
Affiliation(s)
- Dan Song
- Department of Food Science and Engineering, Ningbo University, Ningbo University, Ningbo, P.R. China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo University, Ningbo, P.R. China.,State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, P.R. China
| | - Ying Wang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, P.R. China
| |
Collapse
|
45
|
Abstract
Stress is a nonspecific response of the body to any demand imposed upon it, disrupting the body homoeostasis and manifested with symptoms such as anxiety, depression or even headache. These responses are quite frequent in the present competitive world. The aim of this review is to explore the effect of stress on gut microbiota. First, we summarize evidence of where the microbiota composition has changed as a response to a stressful situation, and thereby the effect of the stress response. Likewise, we review different interventions that can modulate microbiota and could modulate the stress according to the underlying mechanisms whereby the gut-brain axis influences stress. Finally, we review both preclinical and clinical studies that provide evidence of the effect of gut modulation on stress. In conclusion, the influence of stress on gut microbiota and gut microbiota on stress modulation is clear for different stressors, but although the preclinical evidence is so extensive, the clinical evidence is more limited. A better understanding of the mechanism underlying stress modulation through the microbiota may open new avenues for the design of therapeutics that could boost the pursued clinical benefits. These new designs should not only focus on stress but also on stress-related disorders such as anxiety and depression, in both healthy individuals and different populations.
Collapse
|
46
|
Miyakawa M, Kubo S, Oda H, Motoki N, Mizuki M, Tsukahara T, Tanaka M, Yamauchi K, Abe F, Nomiyama T. Effects of Lactoferrin on Sleep Conditions in Children Aged 12-32 Months: A Preliminary, Randomized, Double-Blind, Placebo-Controlled Trial. Nat Sci Sleep 2020; 12:671-677. [PMID: 33061724 PMCID: PMC7532897 DOI: 10.2147/nss.s263106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/19/2020] [Indexed: 01/10/2023] Open
Abstract
PURPOSE To investigate preliminarily the effect of lactoferrin (LF)-fortified formula on sleep conditions in children. STUDY DESIGN A preliminary, randomized, double-blind, placebo-controlled trial. METHODS Healthy children between the ages of 12 and 32 months who attended nursery schools in Japan were divided into two groups and assigned a placebo or LF (48 mg/day)-fortified formula. Children's sleep conditions were investigated before and after the 13-week intervention using the Japanese Sleep Questionnaire for Preschoolers (JSQ-P). RESULTS Altogether, 109 participants were randomized. Eight participants were eliminated due to lost to follow-up, withdrawal of consent, and ineligibility, with 101 participants (placebo, n = 48; LF, n = 53) included in the full analysis set (FAS) and used for analysis. Wake-up time, bedtime, and nighttime sleep were comparable between the two groups before and after intervention. The change in total JSQ-P T scores tended to improve in the LF group (placebo vs LF: 0.5 ± 6.5 vs -1.9 ± 6.1, p = 0.074), in particular, morning symptoms significantly improved (grumpy in the morning, hard to wake-up, and hard to get out of bed) (placebo vs LF: 0.8 ± 6.2 vs -1.9 ± 6.2, p = 0.028). A better trend was also observed in the LF group regarding restless legs syndrome (RLS)-motor (rubs feet at night and touches feet at night) (placebo vs LF: 2.3 ± 10.7 vs -0.6 ± 13.5, p = 0.083) and insufficient sleep (stays up more than one hour later the day before a holiday and wakes up more than one hour later on a holiday) (placebo vs LF: 0.1 ± 9.8 vs -1.7 ± 8.8, p = 0.095). No adverse drug reactions were found. CONCLUSION LF intake may improve sleep condition, especially morning symptoms in children above one year of age.
Collapse
Affiliation(s)
- Momoko Miyakawa
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Shutaro Kubo
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Hirotsugu Oda
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Noriko Motoki
- Department of Preventive Medicine and Public Health, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Masaru Mizuki
- Department of Preventive Medicine and Public Health, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Teruomi Tsukahara
- Department of Preventive Medicine and Public Health, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Miyuki Tanaka
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Koji Yamauchi
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Fumiaki Abe
- Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Tetsuo Nomiyama
- Department of Preventive Medicine and Public Health, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
47
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [PMID: 31460832 DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 2409] [Impact Index Per Article: 401.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
48
|
Besedovsky L, Lange T, Haack M. The Sleep-Immune Crosstalk in Health and Disease. Physiol Rev 2019; 99:1325-1380. [PMID: 30920354 PMCID: PMC6689741 DOI: 10.1152/physrev.00010.2018] [Citation(s) in RCA: 708] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/08/2023] Open
Abstract
Sleep and immunity are bidirectionally linked. Immune system activation alters sleep, and sleep in turn affects the innate and adaptive arm of our body's defense system. Stimulation of the immune system by microbial challenges triggers an inflammatory response, which, depending on its magnitude and time course, can induce an increase in sleep duration and intensity, but also a disruption of sleep. Enhancement of sleep during an infection is assumed to feedback to the immune system to promote host defense. Indeed, sleep affects various immune parameters, is associated with a reduced infection risk, and can improve infection outcome and vaccination responses. The induction of a hormonal constellation that supports immune functions is one likely mechanism underlying the immune-supporting effects of sleep. In the absence of an infectious challenge, sleep appears to promote inflammatory homeostasis through effects on several inflammatory mediators, such as cytokines. This notion is supported by findings that prolonged sleep deficiency (e.g., short sleep duration, sleep disturbance) can lead to chronic, systemic low-grade inflammation and is associated with various diseases that have an inflammatory component, like diabetes, atherosclerosis, and neurodegeneration. Here, we review available data on this regulatory sleep-immune crosstalk, point out methodological challenges, and suggest questions open for future research.
Collapse
Affiliation(s)
- Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Tanja Lange
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Monika Haack
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
49
|
Bannach-Brown A, Tillmann S, MacLeod MR, Wegener G. Administration of galacto-oligosaccharide prebiotics in the Flinders Sensitive Line animal model of depression. BMJ OPEN SCIENCE 2019; 3:e000017. [PMID: 35047681 PMCID: PMC8647602 DOI: 10.1136/bmjos-2018-000017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction Major depressive disorder is the leading source of disability globally and current pharmacological treatments are less than adequate. Animal models such as the Flinders Sensitive Line (FSL) rats are used to mimic aspects of the phenotype in the human disorder and to characterise candidate antidepressant agents. Communication between the gut microbiome and the brain may play an important role in psychiatric disorders such as depression. Interventions targeting the gut microbiota may serve as potential treatments for depression, and this drives increasing research into the effect of probiotics and prebiotics in neuropsychiatric disorders. Prebiotics, galacto-oligosaccharides and fructooligosaccharides that stimulate the activity of gut bacteria have been reported to have a positive impact, reducing anxiety and depressive-like phenotypes and stress-related physiology in mice and rats, as well as in humans. Bimuno, the commercially available beta-galacto-oligosaccharide, has been shown to increase gut microbiota diversity. Aim Here, we aim to investigate the effect of Bimuno on rat anxiety-like and depressive-like behaviour and gut microbiota composition in the FSL model, a genetic model of depression, in comparison to their control, the Flinders Resistant Line (FRL) rats. Methods Sixty-four male rats aged 5–7 weeks, 32 FSL and 32 FRL rats, will be randomised to receive Bimuno or control (4 g/kg) daily for 4 weeks. Animals will be tested by an experimenter unaware of group allocation on the forced swim test to assessed depressive-like behaviour, the elevated plus maze to assess anxiety-like behaviour and the open field test to assess locomotion. Animals will be weighed and food and water intake, per kilogram of bodyweight, will be recorded. Faeces will be collected from each animal prior to the start of the experiment and on the final day to assess the bacterial diversity and relative abundance of bacterial genera in the gut. All outcomes and statistical analysis will be carried out blinded to group allocation, group assignments will be revealed after raw data have been uploaded to Open Science Framework. Two-way analysis of variance will be carried out to investigate the effect of treatment (control or prebiotic) and strain (FSL or FRL) on depressive-like and anxiety-like behaviours.
Collapse
Affiliation(s)
- Alexandra Bannach-Brown
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.,Centre for Research in Evidence-Based Medicine, Bond University, Gold Coast, Australia.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sandra Tillmann
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | | | - Gregers Wegener
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Parkar SG, Kalsbeek A, Cheeseman JF. Potential Role for the Gut Microbiota in Modulating Host Circadian Rhythms and Metabolic Health. Microorganisms 2019; 7:microorganisms7020041. [PMID: 30709031 PMCID: PMC6406615 DOI: 10.3390/microorganisms7020041] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
This article reviews the current evidence associating gut microbiota with factors that impact host circadian-metabolic axis, such as light/dark cycles, sleep/wake cycles, diet, and eating patterns. We examine how gut bacteria possess their own daily rhythmicity in terms of composition, their localization to intestinal niches, and functions. We review evidence that gut bacteria modulate host rhythms via microbial metabolites such as butyrate, polyphenolic derivatives, vitamins, and amines. Lifestyle stressors such as altered sleep and eating patterns that may disturb the host circadian system also influence the gut microbiome. The consequent disruptions to microbiota-mediated functions such as decreased conjugation of bile acids or increased production of hydrogen sulfide and the resultant decreased production of butyrate, in turn affect substrate oxidation and energy regulation in the host. Thus, disturbances in microbiome rhythms may at least partially contribute to an increased risk of obesity and metabolic syndrome associated with insufficient sleep and circadian misalignment. Good sleep and a healthy diet appear to be essential for maintaining gut microbial balance. Manipulating daily rhythms of gut microbial abundance and activity may therefore hold promise for a chrononutrition-based approach to consolidate host circadian rhythms and metabolic homeorhesis.
Collapse
Affiliation(s)
- Shanthi G Parkar
- The New Zealand Institute for Plant & Food Research Limited, Private Bag 11600, Palmerston North 4442, New Zealand.
| | - Andries Kalsbeek
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105BA Amsterdam, The Netherlands.
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - James F Cheeseman
- Department of Anaesthesiology, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|