1
|
Kaur M, Aran KR, Paswan R. A potential role of gut microbiota in stroke: mechanisms, therapeutic strategies and future prospective. Psychopharmacology (Berl) 2024; 241:2409-2430. [PMID: 39463207 DOI: 10.1007/s00213-024-06708-4] [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: 03/19/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
RATIONALE Neurological conditions like Stroke and Alzheimer's disease (AD) often include inflammatory responses in the nervous system. Stroke, linked to high disability and mortality rates, poses challenges related to organ-related complications. Recent focus on understanding the pathophysiology of ischemic stroke includes aspects like cellular excitotoxicity, oxidative stress, cell death mechanisms, and neuroinflammation. OBJECTIVE The objective of this paper is to summarize and explore the pathophysiology of ischemic stroke, elucidates the gut-brain axis mechanism, and discusses recent clinical trials, shedding light on novel treatments and future possibilities. RESULTS Changes in gut architecture and microbiota contribute to dementia by enhancing intestinal permeability, activating the immune system, elevating proinflammatory mediators, altering blood-brain barrier (BBB) permeability, and ultimately leading to neurodegenerative diseases (NDDs). The gut-brain axis's potential role in disease pathophysiology offers new avenues for cell-based regenerative medicine in treating neurological conditions. CONCLUSION In conclusion, the gut microbiome significantly impacts stroke prognosis by highlighting the role of the gut-brain axis in ischemic stroke mechanisms. This insight suggests potential therapeutic strategies for improving outcomes.
Collapse
Affiliation(s)
- Manpreet Kaur
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Raju Paswan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
2
|
Qian X, Li Q, Zhu H, Chen Y, Lin G, Zhang H, Chen W, Wang G, Tian P. Bifidobacteria with indole-3-lactic acid-producing capacity exhibit psychobiotic potential via reducing neuroinflammation. Cell Rep Med 2024; 5:101798. [PMID: 39471819 PMCID: PMC11604549 DOI: 10.1016/j.xcrm.2024.101798] [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: 02/28/2024] [Revised: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 11/01/2024]
Abstract
The escalating global prevalence of depression demands effective therapeutic strategies, with psychobiotics emerging as a promising solution. However, the molecular mechanisms governing the neurobehavioral impact of psychobiotics remain elusive. This study reveals a significant reduction in hippocampal indole-3-lactic acid (ILA) levels in depressed mice, which is ameliorated by the psychobiotic Bifidobacterium breve. In both human subjects and mice, the ILA increase in the circulatory system results from bifidobacteria supplementation. Further investigation identifies the key aromatic lactate dehydrogenase (Aldh) gene and pathway in bifidobacteria responsible for ILA production. Importantly, the antidepressant effects are nullified in the Aldh mutants compared to the wild-type strain. At the bifidobacteria species level, those with Aldh exhibit heightened antidepressant effects. Finally, this study emphasizes the antidepressant efficacy of psychobiotic-derived ILA, potentially mediated by aryl hydrocarbon receptor (AhR) signaling activation to alleviate neuroinflammation. This study unveils the molecular and genetic foundations of psychobiotics' antidepressant effects, offering insights for microbial therapies targeting mood disorders.
Collapse
Affiliation(s)
- Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qing Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guopeng Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
3
|
Jain M, Anand A, Sharma N, Shamim MA, Enioutina EY. Effect of Probiotics Supplementation on Cortisol Levels: A Systematic Review and Meta-Analysis. Nutrients 2024; 16:3564. [PMID: 39458560 PMCID: PMC11510182 DOI: 10.3390/nu16203564] [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: 08/31/2024] [Revised: 09/25/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Several randomized controlled trials (RCTs) have shown conflicting results on cortisol levels following probiotic administration in healthy and diseased populations. Previous analyses were inconclusive due to limited studies, and evidence is lacking on how these effects vary by health status; region; therapy duration; medications, and use of single or multiple strains. Methods: In this systematic review and meta-analysis (PROSPERO [CRD42024538539]), we searched PubMed, Cochrane Library, Embase, Scopus, Web of Science, CINAHL, ProQuest, and Web of Science Preprints until 13 August 2024, for RCTs on probiotic administration, either alone or combined, across all age groups and without specific medical condition requirements. We applied random-effects meta-analysis, assessed bias using the Cochrane RoB 2 tool, and evaluated evidence certainty with GRADE. Findings: We screened 1739 records and retrieved 46 RCTs (3516 participants). Probiotics supplementation decreased cortisol levels compared to the control arm [46 RCTs; SMD: -0.45; 95% CI: -0.83; -0.07; I2: 92.5%, low certainty]. Among various subgroups; probiotics supplementation decreased the cortisol levels in the subgroups without concomitant medications [37 RCTs; SMD: -0.30; 95% CI [-0.58; -0.03], I2: 88.7%] with a single probiotic strain [30 RCTs; SMD: -0.33; 95% CI: -0.63; -0.028; I2: 88.8%], in a healthy population [35 RCTs; SMD:-0.3; 95% CI: -0.58; -0.03; I2: 88.7] and in the Asia region [21 RCTs; SMD: -0.83; 95% CI: -1.58; -0.07; I2: 95%]. Interpretation: A low level of evidence suggests probiotics might reduce cortisol levels, but more targeted studies are needed to identify variables affecting the response in specific subgroups.
Collapse
Affiliation(s)
- Manav Jain
- Division of Clinical Pharmacology, Department of Pediatrics, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT 84108, USA;
| | - Aishwarya Anand
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Nisha Sharma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India;
| | - Muhammad Aaqib Shamim
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur 342005, India;
| | - Elena Y. Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT 84108, USA;
| |
Collapse
|
4
|
Santamarina AB, Nehmi Filho V, Freitas JAD, Franco LAM, Fonseca JV, Martins RC, Turri JAO, Silva BFRBD, Gusmão AF, Olivieri EHR, Otoch JP, Pessoa AFM. Nutraceutical Capsules LL1 and Silymarin Supplementation Act on Mood and Sleep Quality Perception by Microbiota-Gut-Brain Axis: A Pilot Clinical Study. Nutrients 2024; 16:3049. [PMID: 39339649 PMCID: PMC11435014 DOI: 10.3390/nu16183049] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut-brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study aimed to investigate a new nutraceutical formulation comprising prebiotics, minerals, and silymarin's impact on microbiota, inflammation, mood, and sleep quality. The study evaluated the LL1 + silymarin capsule supplementation over 180 days in overweight adults. We analyzed the fecal gut microbiota using partial 16S rRNA sequences, measured cytokine expression via CBA, collected anthropometric data, quality of life, and sleep questionnaire responses, and obtained plasma samples for metabolic and hormonal analysis at baseline (T0) and 180 days (T180) post-supplementation. Our findings revealed significant reshaping in gut microbiota composition at the phylum, genus, and species levels, especially in the butyrate-producer bacteria post-supplementation. These changes in gut microbiota were linked to enhancements in sleep quality, mood perception, cytokine expression, and anthropometric measures which microbiota-derived short-chain fatty acids might enhance. The supplementation tested in this study seems to be able to improve microbiota composition, reflecting anthropometrics and inflammation, as well as sleep quality and mood improvement.
Collapse
Affiliation(s)
- Aline Boveto Santamarina
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo 03317-000, SP, Brazil
| | - Victor Nehmi Filho
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo 03317-000, SP, Brazil
| | - Jéssica Alves de Freitas
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo 03317-000, SP, Brazil
| | - Lucas Augusto Moysés Franco
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Joyce Vanessa Fonseca
- Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM-49), Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Roberta Cristina Martins
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - José Antônio Orellana Turri
- Grupo de Pesquisa em Economia da Saúde, Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
| | - Bruna Fernanda Rio Branco da Silva
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Laboratório Interdisciplinar em Fisiologia e Exercício, Universidade Federal de São Paulo (UNIFESP), Santos 11015-020, SP, Brazil
| | - Arianne Fagotti Gusmão
- International Research Center, A.C. Camargo Cancer Center, São Paulo 01508-010, SP, Brazil
| | | | - José Pinhata Otoch
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Hospital Universitário da Universidade de São Paulo, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina de São Paulo, Universidade de São Paulo, São Paulo 01246-903, SP, Brazil
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| |
Collapse
|
5
|
Zarimeidani F, Rahmati R, Mostafavi M, Darvishi M, Khodadadi S, Mohammadi M, Shamlou F, Bakhtiyari S, Alipourfard I. Gut Microbiota and Autism Spectrum Disorder: A Neuroinflammatory Mediated Mechanism of Pathogenesis? Inflammation 2024:10.1007/s10753-024-02061-y. [PMID: 39093342 DOI: 10.1007/s10753-024-02061-y] [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: 01/31/2024] [Revised: 04/28/2024] [Accepted: 05/21/2024] [Indexed: 08/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social communication and behavior, frequently accompanied by restricted and repetitive patterns of interests or activities. The gut microbiota has been implicated in the etiology of ASD due to its impact on the bidirectional communication pathway known as the gut-brain axis. However, the precise involvement of the gut microbiota in the causation of ASD is unclear. This study critically examines recent evidence to rationalize a probable mechanism in which gut microbiota symbiosis can induce neuroinflammation through intermediator cytokines and metabolites. To develop ASD, loss of the integrity of the intestinal barrier, activation of microglia, and dysregulation of neurotransmitters are caused by neural inflammatory factors. It has emphasized the potential role of neuroinflammatory intermediates linked to gut microbiota alterations in individuals with ASD. Specifically, cytokines like brain-derived neurotrophic factor, calprotectin, eotaxin, and some metabolites and microRNAs have been considered etiological biomarkers. We have also overviewed how probiotic trials may be used as a therapeutic strategy in ASD to reestablish a healthy balance in the gut microbiota. Evidence indicates neuroinflammation induced by dysregulated gut microbiota in ASD, yet there is little clarity based on analysis of the circulating immune profile. It deems the repair of microbiota load would lower inflammatory chaos in the GI tract, correct neuroinflammatory mediators, and modulate the neurotransmitters to attenuate autism. The interaction between the gut and the brain, along with alterations in microbiota and neuroinflammatory biomarkers, serves as a foundational background for understanding the etiology, diagnosis, prognosis, and treatment of autism spectrum disorder.
Collapse
Affiliation(s)
- Fatemeh Zarimeidani
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rahem Rahmati
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehrnaz Mostafavi
- Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Darvishi
- School of Aerospace and Subaquatic Medicine, Infectious Diseases & Tropical Medicine Research Center (IDTMC), AJA University of Medical Sciences, Tehran, Iran
| | - Sanaz Khodadadi
- Student Research Committee, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Shamlou
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Salar Bakhtiyari
- Feinberg Cardiovascular and Renal Research Institute, North Western University, Chicago. Illinois, USA
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcin Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
6
|
Chaisanam R, Wattanathorn J, Thukham-mee W, Piyavhatkul N, Paholpak P. Anxiolytic, Antidepression, and Memory-Enhancing Effects of the Novel Instant Soup RJ6601 in the Middle-Aged of Female Rats. Foods 2024; 13:2170. [PMID: 39063255 PMCID: PMC11276534 DOI: 10.3390/foods13142170] [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: 06/08/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Due to the health benefits of polyphenols and dietary fiber in combating mental disorders, we hypothesized that a polyphenol- and dietary fiber-enriched soup (RJ6601) would improve mental wellness in a rat model of middle-aged women. To test this hypothesis, female Wistar rats aged 18 months (350-450 g) were orally administered RJ6601 at doses of 200 and 400 mg/kg BW for 28 days. The anxiolytic, antidepression, and memory-enhancing effects were assessed every 7 days throughout the study period. The neuron density and levels of activities of AChE, total MAO, MAO-A, MAO-B, MDA, SOD, CAT, GSH-Px, IL-1β, IL-6, and BDNF in the prefrontal cortex at the end of study were also investigated. Furthermore, the amounts of Lactobacillus spp. and Bifidobacterium spp. in their feces were also determined. The results revealed that the developed soup shows anxiolytic, antidepression, and memory-enhancing effects. An increased neuron density; reductions in AChE, total MAO, MAO-A, MAO-B, and MDA; and an elevation of serum BDNF, together with increased amounts of both bacterial species in feces, were also observed. Our results suggest that RJ6601 is a potential mental wellness promotion supplement that enhances BDNF levels, brain plasticity, neurotransmitter balance, and oxidative stress and inflammation status, along with improving microbiota.
Collapse
Affiliation(s)
- Rujikan Chaisanam
- Department of Physiology and Graduate School (Neuroscience Program), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Jintanaporn Wattanathorn
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Research Institute for High Human Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wipawee Thukham-mee
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Research Institute for High Human Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nawanant Piyavhatkul
- Department Psychiatry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (N.P.); (P.P.)
| | - Pongsatorn Paholpak
- Department Psychiatry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (N.P.); (P.P.)
| |
Collapse
|
7
|
Xie Y, Zhu H, Yuan Y, Guan X, Xie Q, Dong Z. Baseline gut microbiota profiles affect treatment response in patients with depression. Front Microbiol 2024; 15:1429116. [PMID: 39021622 PMCID: PMC11251908 DOI: 10.3389/fmicb.2024.1429116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The role of the gut microbiota in the pathophysiology of depression has been explored in numerous studies, which have confirmed that the baseline gut microbial profiles of patients with depression differ from those of healthy individuals. The gut microbiome affects metabolic activity in the immune and central nervous systems and regulates intestinal ecology through the neuroendocrine system. Additionally, baseline changes in the gut microbiota differed among patients with depression who demonstrated varying treatment response. Currently, probiotics are an emerging treatment for depression; however, the efficacy of modulating the gut microbiota in the treatment of depression remains uncertain. Additionally, the mechanisms by which changes in the gut microbiota affect treatment response in patients with depression remain unclear. In this review, we aimed to summarize the differences in the baseline gut microbiota between the remission and non-remission groups after antidepressant therapy. Additionally, we summarized the possible mechanisms that may contribute to antidepressant resistance through the effects of the gut microbiome on the immune and nervous systems, various enzymes, bioaccumulation, and blood-brain barrier, and provide a basis for treating depression by targeting the gut microbiota.
Collapse
Affiliation(s)
- Yingjing Xie
- West China Hospital, Sichuan University, Chengdu, China
| | - Hanwen Zhu
- West China Hospital, Sichuan University, Chengdu, China
| | - Yanling Yuan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xuan Guan
- Chengdu Medical College, Chengdu, China
| | - Qinglian Xie
- Department of Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Zaiquan Dong
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Kim N, Parolin B, Renshaw D, Deb SK, Zariwala MG. Formulated Palmitoylethanolamide Supplementation Improves Parameters of Cognitive Function and BDNF Levels in Young, Healthy Adults: A Randomised Cross-Over Trial. Nutrients 2024; 16:489. [PMID: 38398813 PMCID: PMC10891801 DOI: 10.3390/nu16040489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Palmitoylethanolamide (PEA) is an endocannabinoid-like lipid mediator which is naturally produced in the body and found in certain foods. The aim of this study was to assess the effect of a bioavailable formulated form of PEA (Levagen+®) on serum BDNF levels and parameters of cognitive function in healthy adults. METHODS A randomised double-blinded placebo-controlled cross-over trial was implemented to measure the effects of a 6-week 700 mg/day course of formulated PEA supplementation versus a placebo. Participants (n = 39) completed pre- and post-assessments of a lab-based cognitive test. Serum samples were collected to measure BDNF concentrations using an immunoassay. RESULTS A significant increase in serum BDNF levels was found following PEA supplementation compared with the placebo (p = 0. 0057, d = 0.62). The cognition test battery demonstrated improved memory with PEA supplementation through better first success (p = 0.142, d = 0.54) and fewer errors (p = 0.0287; d = -0.47) on the Paired Associates Learning test. CONCLUSION This was the first study to report a direct beneficial effect of Levagen+® PEA supplementation on memory improvement as well as corresponding increases in circulating neurotrophic marker levels. This suggests that formulated PEA holds promise as an innovative and practical intervention for cognitive health enhancement.
Collapse
Affiliation(s)
- Nadia Kim
- Centre for Nutraceuticals, University of Westminster, London W1W6 UW, UK (S.K.D.)
| | - Brenda Parolin
- Centre for Nutraceuticals, University of Westminster, London W1W6 UW, UK (S.K.D.)
| | - Derek Renshaw
- Centre for Health and Life Sciences (CHLS), Coventry University, Coventry CV1 5FB, UK;
| | - Sanjoy K. Deb
- Centre for Nutraceuticals, University of Westminster, London W1W6 UW, UK (S.K.D.)
- Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK
| | | |
Collapse
|
9
|
Mishra V, Yadav D, Solanki KS, Koul B, Song M. A Review on the Protective Effects of Probiotics against Alzheimer's Disease. BIOLOGY 2023; 13:8. [PMID: 38248439 PMCID: PMC10813289 DOI: 10.3390/biology13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
This review summarizes the protective effects of probiotics against Alzheimer's disease (AD), one of the most common neurodegenerative disorders affecting older adults. This disease is characterized by the deposition of tau and amyloid β peptide (Aβ) in different parts of the brain. Symptoms observed in patients with AD include struggles with writing, speech, memory, and knowledge. The gut microbiota reportedly plays an important role in brain functioning due to its bidirectional communication with the gut via the gut-brain axis. The emotional and cognitive centers in the brain are linked to the functions of the peripheral intestinal system via this gut-brain axis. Dysbiosis has been linked to neurodegenerative disorders, indicating the significance of gut homeostasis for proper brain function. Probiotics play an important role in protecting against the symptoms of AD as they restore gut-brain homeostasis to a great extent. This review summarizes the characteristics, status of gut-brain axis, and significance of gut microbiota in AD. Review and research articles related to the role of probiotics in the treatment of AD were searched in the PubMed database. Recent studies conducted using animal models were given preference. Recent clinical trials were searched for separately. Several studies conducted on animal and human models clearly explain the benefits of probiotics in improving cognition and memory in experimental subjects. Based on these studies, novel therapeutic approaches can be designed for the treatment of patients with AD.
Collapse
Affiliation(s)
- Vibhuti Mishra
- School of Studies in Biochemistry, Jiwaji University, Gwalior 474003, India;
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Kavita Singh Solanki
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA;
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India;
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
10
|
Hsu YC, Huang YY, Tsai SY, Kuo YW, Lin JH, Ho HH, Chen JF, Hsia KC, Sun Y. Efficacy of Probiotic Supplements on Brain-Derived Neurotrophic Factor, Inflammatory Biomarkers, Oxidative Stress and Cognitive Function in Patients with Alzheimer's Dementia: A 12-Week Randomized, Double-Blind Active-Controlled Study. Nutrients 2023; 16:16. [PMID: 38201846 PMCID: PMC10780998 DOI: 10.3390/nu16010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
The role of neurotrophic factors, oxidative stress, and inflammation in the pathogenesis of Alzheimer's disease (AD) has been explored. Animal studies have reported the positive effects of probiotics on these factors. Some clinical studies also support the potential role of probiotics in improving cognitive function via the gut-brain axis in older adults. However, clinical experimental studies evaluating the efficacy of probiotics targeting the neurotrophic factors and inflammatory biomarkers, particularly among AD patients, remain very limited. In this randomized, double-blinded, active-controlled trial, we used multi-strain probiotic supplements, including Bifidobacterium longum subsp. infantis BLI-02, B. breve Bv-889, B. animalis subsp. lactis CP-9, B. bifidum VDD088, and Lactobacillus plantarum PL-02 as the intervention. Participants were divided into an active control group (received probiotic supplements containing 5 × 107 colony-forming units per day, CFU/day) and a treatment group (1 × 1010 CFU/day). Student's t test was applied as the main method of statistical analysis. After 12 weeks of intervention, the treatment group demonstrated a 36% increase in serum brain-derived neurotrophic factor (BDNF) (* p = 0.005), a reduction in IL-1β (* p = 0.041), and an increase in antioxidant superoxide dismutase (SOD) (* p = 0.012). No significant change was found in the active control group. A trend toward less cognitive deterioration was observed, but not statistically significant. In conclusion, this study presents evidence supporting the benefits of multi-strain probiotics in enhancing BDNF, ameliorating inflammation and oxidative stress in AD patients.
Collapse
Affiliation(s)
- Yu-Chieh Hsu
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Yen-Yu Huang
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Shin-Yu Tsai
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Yi-Wei Kuo
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Jia-Hung Lin
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Hsieh-Hsun Ho
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Jui-Fen Chen
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Ko-Chiang Hsia
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (Y.-W.K.); (J.-H.L.); (H.-H.H.); (J.-F.C.); (K.-C.H.)
| | - Yu Sun
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
11
|
Kujawa D, Laczmanski L, Budrewicz S, Pokryszko-Dragan A, Podbielska M. Targeting gut microbiota: new therapeutic opportunities in multiple sclerosis. Gut Microbes 2023; 15:2274126. [PMID: 37979154 PMCID: PMC10730225 DOI: 10.1080/19490976.2023.2274126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/18/2023] [Indexed: 11/20/2023] Open
Abstract
Multiple sclerosis (MS) causes long-lasting, multifocal damage to the central nervous system. The complex background of MS is associated with autoimmune inflammation and neurodegeneration processes, and is potentially affected by many contributing factors, including altered composition and function of the gut microbiota. In this review, current experimental and clinical evidence is presented for the characteristics of gut dysbiosis found in MS, as well as for its relevant links with the course of the disease and the dysregulated immune response and metabolic pathways involved in MS pathology. Furthermore, therapeutic implications of these investigations are discussed, with a range of pharmacological, dietary and other interventions targeted at the gut microbiome and thus intended to have beneficial effects on the course of MS.
Collapse
Affiliation(s)
- Dorota Kujawa
- Laboratory of Genomics & Bioinformatics, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Lukasz Laczmanski
- Laboratory of Genomics & Bioinformatics, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | | | | | - Maria Podbielska
- Laboratory of Microbiome Immunobiology, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
12
|
Azuma N, Mawatari T, Saito Y, Tsukamoto M, Sampei M, Iwama Y. Effect of Continuous Ingestion of Bifidobacteria and Dietary Fiber on Improvement in Cognitive Function: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:4175. [PMID: 37836458 PMCID: PMC10574581 DOI: 10.3390/nu15194175] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Bifidobacterium animalis subsp. lactis GCL2505 has been shown to have some positive effects on health, including improved defecation frequency and reduced visceral fat. These effects are thought to be due to GCL2505's unique ability to reach the intestine in a viable form and proliferate after a single intake. This leads to an increased number of intestinal bifidobacteria. This randomized, double-blind, placebo-controlled, parallel-group study was conducted to confirm that intake of GCL2505 and inulin (a prebiotic) improve cognitive function (n = 80). Participants consumed test drinks containing 1 × 1010 colony-forming units of GCL2505 per 100 g and 2.0 g of inulin per 100 g for 12 weeks. The change in cognitive function assessment scores was set as the primary endpoint. There were significant improvements in scores in the neurocognitive index domain, which is an assessment of overall cognitive function, in addition to overall attention, cognitive flexibility, and executive function domains. The intervention significantly increased the number of fecal bifidobacteria and affected the levels of several inflammatory markers. These results suggest that intake of GCL2505 and inulin improves cognitive function by improving the intestinal environment and alleviating inflammation.
Collapse
Affiliation(s)
- Naoki Azuma
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Takashi Mawatari
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Yasuo Saito
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Masashi Tsukamoto
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Masatoshi Sampei
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Yoshitaka Iwama
- Nihonbashi Cardiology Clinic, Kyodo Bldg. #201, 13-4 Nihonbashi Kodenmacho, Chuo-Ku, Tokyo 103-0001, Japan;
| |
Collapse
|
13
|
Di Gesù CM, Matz LM, Fultz R, Bolding IJ, Buffington SA. Monospecies probiotic preparation and administration with downstream analysis of sex-specific effects on gut microbiome composition in mice. STAR Protoc 2023; 4:102386. [PMID: 37379217 PMCID: PMC10331592 DOI: 10.1016/j.xpro.2023.102386] [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: 02/06/2023] [Revised: 04/16/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
Dysbiosis of the gut microbiome is implicated in the growing burden of non-communicable chronic diseases, including neurodevelopmental disorders, and both preclinical and clinical studies highlight the potential for precision probiotic therapies in their prevention and treatment. Here, we present an optimized protocol for the preparation and administration of Limosilactobacillus reuteri MM4-1A (ATCC-PTA-6475) to adolescent mice. We also describe steps for performing downstream analysis of metataxonomic sequencing data with careful assessment of sex-specific effects on microbiome composition and structure. For complete details on the use and execution of this protocol, please refer to Di Gesù et al.1.
Collapse
Affiliation(s)
- Claudia M Di Gesù
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy
| | - Lisa M Matz
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Robert Fultz
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Ian J Bolding
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Shelly A Buffington
- Department of Neurobiology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Sealy Center for Microbiome Research, The University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
14
|
Li Y, Tong T, Li P, Peng Y, Zhang M, Liu J, She Y, Li Z, Li Y. Screening of Potential Probiotic Lactobacillaceae and Their Improvement of Type 2 Diabetes Mellitus by Promoting PI3K/AKT Signaling Pathway in db/db Mice. Pol J Microbiol 2023; 72:285-297. [PMID: 37725896 PMCID: PMC10508973 DOI: 10.33073/pjm-2023-028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/09/2023] [Indexed: 09/21/2023] Open
Abstract
The study aimed to isolate Lactobacillaceae strains with in vitro hypoglycemic activity and probiotic properties and to determine their antidiabetic abilities in vivo. Lactiplantibacillus plantarum 22, L. plantarum 25, Limosilactobacillus fermentum 11, and L. fermentum 305 with high in vitro hypoglycemic activity were screened from 23 strains of Lactobacillaceae isolated from human feces and identified by 16S rDNA sequencing. The fasting blood glucose (FBG) of the mice was recorded weekly. After 12 weeks, liver, kidney, and pancreas tissues were stained with hematoxylin and eosin (H&E) to observe histomorphology; the inflammatory factors were assayed by Quantitative Real-time PCR; PI3K and AKT were measured by Western blot; the short-chain fatty acids (SCFAs) were determined by LC-MS/MS. Inhibitory activities of L. plantarum 22, L. plantarum 25, L. fermentum 11, and L. fermentum 305 against α-amylase were 62.29 ± 0.44%, 51.81 ± 3.65%, 58.40 ± 1.68%, and 57.48 ± 5.04%, respectively. Their inhibitory activities to α-glucosidase were 14.89 ± 0.38%, 15.32 ± 0.89%, 52.63 ± 3.07%, and 51.79 ± 1.13%, respectively. Their survival rate after simulated gastrointestinal test were 12.42 ± 2.84%, 9.10 ± 1.12%, 5.86 ± 0.52%, and 8.82 ± 2.50% and their adhesion rates to Caco-2 cell were 6.09 ± 0.39%, 6.37 ± 0.28%, 6.94 ± 0.27%, and 6.91 ± 0.11%, respectively. The orthogonal tests of bacterial powders of the four strains showed that the maximum inhibitory activities to α-amylase and α-glucosidase were 93.18 ± 1.19% and 75.33 ± 2.89%, respectively. The results showed that the mixture of Lactobacillaceae could lower FBG, reduce inflammation, and liver, kidney, and pancreas damage, promote PI3K/AKT signaling pathway, and increase the content of SCFAs. The combination of L. plantarum 22, L. plantarum 25, L. fermentum 11, and L. fermentum 305 can potentially improve type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Yueyang Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Tong Tong
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Peifan Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yian Peng
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- School of Public Health, Anhui University of Science and Technology, Hefei, China
| | - Jia Liu
- Internal Trade Food Science and Technology (Beijing) Co., Ltd., Beijing, China
| | - Yongxin She
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Zuming Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yongli Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Vitetta L, Bambling M, Strodl E. Probiotics and Commensal Bacteria Metabolites Trigger Epigenetic Changes in the Gut and Influence Beneficial Mood Dispositions. Microorganisms 2023; 11:1334. [PMID: 37317308 DOI: 10.3390/microorganisms11051334] [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: 05/05/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023] Open
Abstract
The effect of the intestinal microbiome on the gut-brain axis has received considerable attention, strengthening the evidence that intestinal bacteria influence emotions and behavior. The colonic microbiome is important to health and the pattern of composition and concentration varies extensively in complexity from birth to adulthood. That is, host genetics and environmental factors are complicit in shaping the development of the intestinal microbiome to achieve immunological tolerance and metabolic homeostasis from birth. Given that the intestinal microbiome perseveres to maintain gut homeostasis throughout the life cycle, epigenetic actions may determine the effect on the gut-brain axis and the beneficial outcomes on mood. Probiotics are postulated to exhibit a range of positive health benefits including immunomodulating capabilities. Lactobacillus and Bifidobacterium are genera of bacteria found in the intestines and so far, the benefits afforded by ingesting bacteria such as these as probiotics to people with mood disorders have varied in efficacy. Most likely, the efficacy of probiotic bacteria at improving mood has a multifactorial dependency, relying namely on several factors that include the agents used, the dose, the pattern of dosing, the pharmacotherapy used, the characteristics of the host and the underlying luminal microbial environment (e.g., gut dysbiosis). Clarifying the pathways linking probiotics with improvements in mood may help identify the factors that efficacy is dependent upon. Adjunctive therapies with probiotics for mood disorders could, through DNA methylation molecular mechanisms, augment the intestinal microbial active cohort and endow its mammalian host with important and critical co-evolutionary redox signaling metabolic interactions, that are embedded in bacterial genomes, and that in turn can enhance beneficial mood dispositions.
Collapse
Affiliation(s)
- Luis Vitetta
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2005, Australia
| | - Matthew Bambling
- Faculty of Medicine and Health, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Esben Strodl
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4058, Australia
| |
Collapse
|
16
|
Gao J, Zhao L, Cheng Y, Lei W, Wang Y, Liu X, Zheng N, Shao L, Chen X, Sun Y, Ling Z, Xu W. Probiotics for the treatment of depression and its comorbidities: A systemic review. Front Cell Infect Microbiol 2023; 13:1167116. [PMID: 37139495 PMCID: PMC10149938 DOI: 10.3389/fcimb.2023.1167116] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 05/05/2023] Open
Abstract
Depression is one of the most common psychiatric conditions, characterized by significant and persistent depressed mood and diminished interest, and often coexists with various comorbidities. The underlying mechanism of depression remain elusive, evidenced by the lack of an appreciate therapy. Recent abundant clinical trials and animal studies support the new notion that the gut microbiota has emerged as a novel actor in the pathophysiology of depression, which partakes in bidirectional communication between the gut and the brain through the neuroendocrine, nervous, and immune signaling pathways, collectively known as the microbiota-gut-brain (MGB) axis. Alterations in the gut microbiota can trigger the changes in neurotransmitters, neuroinflammation, and behaviors. With the transition of human microbiome research from studying associations to investigating mechanistic causality, the MGB axis has emerged as a novel therapeutic target in depression and its comorbidities. These novel insights have fueled idea that targeting on the gut microbiota may open new windows for efficient treatment of depression and its comorbidities. Probiotics, live beneficial microorganisms, can be used to modulate gut dysbiosis into a new eubiosis and modify the occurrence and development of depression and its comorbidities. In present review, we summarize recent findings regarding the MGB axis in depression and discuss the potential therapeutic effects of probiotics on depression and its comorbidities.
Collapse
Affiliation(s)
- Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yu Wang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nengneng Zheng
- Department of Obstetrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xulei Chen
- Department of Psychiatry, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Yilai Sun
- Department of Psychiatry, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Weijie Xu
- Department of Psychiatry, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
17
|
Karbownik MS, Sokołowska P, Kowalczyk E. Gut Microbiota Metabolites Differentially Release Gliotransmitters from the Cultured Human Astrocytes: A Preliminary Report. Int J Mol Sci 2023; 24:ijms24076617. [PMID: 37047602 PMCID: PMC10095279 DOI: 10.3390/ijms24076617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Butyrate and indole-3-propionic acid represent the CNS-available gut microbiota metabolites exhibiting potentially beneficial effects on human brain function and being tested as antidepressants. Astrocytes represent one of the putative targets for the gut metabolites; however, the mechanism of action of butyrate and indole-3-propionic acid is not well understood. In order to test this mechanism, a human astrocyte cell-line culture was treated with the compounds or without them, and the supernatants were collected for the analysis of ATP and glutamate gliotransmitter release with the use of luminescent and fluorescent methods, respectively. A 10-min incubation of astrocytes with 1–5 mM butyrate increased the ATP gliotransmitter release by 78% (95%CI: 45–119%), p < 0.001. The effect was found to be mediated by the cytosolic Ca2+ mobilization. Both 10-min and 24-h treatments with indole-3-propionic acid produced no significant effects on the release of gliotransmitters. The results for glutamate release were inconclusive due to a specific glutamate release pattern discovered in the tested model. This preliminary report of butyrate-induced ATP gliotransmitter release appears to provide a novel mechanistic explanation for the beneficial effect of this gut microbiota metabolite on brain function; however, the results require further evaluation in more composed models.
Collapse
Affiliation(s)
- Michał Seweryn Karbownik
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
18
|
Johnson D, Letchumanan V, Thum CC, Thurairajasingam S, Lee LH. A Microbial-Based Approach to Mental Health: The Potential of Probiotics in the Treatment of Depression. Nutrients 2023; 15:nu15061382. [PMID: 36986112 PMCID: PMC10053794 DOI: 10.3390/nu15061382] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are currently the subject of intensive research pursuits and also represent a multi-billion-dollar global industry given their vast potential to improve human health. In addition, mental health represents a key domain of healthcare, which currently has limited, adverse-effect prone treatment options, and probiotics may hold the potential to be a novel, customizable treatment for depression. Clinical depression is a common, potentially debilitating condition that may be amenable to a precision psychiatry-based approach utilizing probiotics. Although our understanding has not yet reached a sufficient level, this could be a therapeutic approach that can be tailored for specific individuals with their own unique set of characteristics and health issues. Scientifically, the use of probiotics as a treatment for depression has a valid basis rooted in the microbiota-gut-brain axis (MGBA) mechanisms, which play a role in the pathophysiology of depression. In theory, probiotics appear to be ideal as adjunct therapeutics for major depressive disorder (MDD) and as stand-alone therapeutics for mild MDD and may potentially revolutionize the treatment of depressive disorders. Although there is a wide range of probiotics and an almost limitless range of therapeutic combinations, this review aims to narrow the focus to the most widely commercialized and studied strains, namely Lactobacillus and Bifidobacterium, and to bring together the arguments for their usage in patients with major depressive disorder (MDD). Clinicians, scientists, and industrialists are critical stakeholders in exploring this groundbreaking concept.
Collapse
Affiliation(s)
- Dinyadarshini Johnson
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Vengadesh Letchumanan
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Chern Choong Thum
- Department of Psychiatry, Hospital Sultan Abdul Aziz Shah, Persiaran Mardi-UPM, Serdang 43400, Malaysia
| | - Sivakumar Thurairajasingam
- Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| |
Collapse
|
19
|
Xu F, Xie Q, Kuang W, Dong Z. Interactions Between Antidepressants and Intestinal Microbiota. Neurotherapeutics 2023; 20:359-371. [PMID: 36881351 PMCID: PMC10121977 DOI: 10.1007/s13311-023-01362-8] [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] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
The microbiota-gut-brain axis has been shown to influence human health and diseases, including depression. The interactions between drugs and intestinal microbiota are complex and highly relevant to treat diseases. Studies have shown an interaction between antidepressants and intestinal microbiota. Antidepressants may alter the abundance and composition of intestinal microbiota, which are closely related to the treatment outcomes of depression. Intestinal microbiota can influence the metabolism of antidepressants to change their availability (e.g., tryptophan can be metabolized to kynurenine by intestinal microbiota) and regulate their absorption by affecting intestinal permeability. In addition, the permeability of the blood-brain barrier can be altered by intestinal microbiota, influencing antidepressants to reach the central nervous system. Bioaccumulation is also a type of drug-microbiota interaction, which means bacteria accumulate drugs without biotransformation. These findings imply that it is important to consider intestinal microbiota when evaluating antidepressant therapy regimens and that intestinal microbiota can be a potential target for depression treatment.
Collapse
Affiliation(s)
- Feiyu Xu
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Qinglian Xie
- Department of Outpatient, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weihong Kuang
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaiquan Dong
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
20
|
Peripheral Regulation of Central Brain-Derived Neurotrophic Factor Expression through the Vagus Nerve. Int J Mol Sci 2023; 24:ijms24043543. [PMID: 36834953 PMCID: PMC9964523 DOI: 10.3390/ijms24043543] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The brain-derived neurotrophic factor (BDNF) is an extensively studied neurotrophin es sential for both developing the brain and maintaining adult brain function. In the adult hippocampus, BDNF is critical for maintaining adult neurogenesis. Adult hippocampal neurogenesis is involved not only in memory formation and learning ability, but also mood regulation and stress responses. Accordingly, decreased levels of BDNF, accompanied by low levels of adult neurogenesis, occurs in brains of older adults with impaired cognitive function and in those of patients with major depression disorder. Therefore, elucidating the mechanisms that maintain hippocampal BDNF levels is biologically and clinically important. It has been revealed that signalling from peripheral tissues contribute to the regulation of BDNF expression in the brain across the blood-brain barrier. Moreover, recent studies indicated evidence that neuronal pathways can also be a mechanism by which peripheral tissues signal to the brain for the regulation of BDNF expression. In this review, we give an overview of the current status in the regulation of central BDNF expression by peripheral signalling, with a special interest in the regulation of hippocampal BDNF levels by signals via the vagus nerve. Finally, we discuss the relationship between signalling from peripheral tissues and age-associated control of central BDNF expression.
Collapse
|
21
|
Hua H, Huang C, Liu H, Xu X, Xu X, Wu Z, Liu C, Wang Y, Yang C. Depression and antidepressant effects of ketamine and its metabolites: The pivotal role of gut microbiota. Neuropharmacology 2022; 220:109272. [PMID: 36170927 DOI: 10.1016/j.neuropharm.2022.109272] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 02/07/2023]
Abstract
The discovery of the robust antidepressant actions of ketamine is regarded as one of the greatest advancements in depression treatment in the past 60 years. Recent findings have provided strong evidence for the presence of bidirectional communication networks between the gastrointestinal tract and the brain in depression. Moreover, increasing evidence supports the antidepressant role of ketamine in regulating the gut microbiome and microbiota-derived molecules; however, the mechanisms underpinning such effects are still ambiguous. This review summarizes the current understanding of the anti-depressant mechanisms of ketamine and its metabolites regarding the bidirectional regulation by microbiota-gut-brain axis. We review the relationship between gut microbiota and the antidepressant mechanisms of ketamine, and discuss the role of stress response, brain-derived neurotrophic factor (BDNF)-mediated neurogenesis, anti-inflammatory effect and neurotransmitters.
Collapse
Affiliation(s)
- Hao Hua
- Department of Anesthesiology, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, 214062, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hanyu Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiangyang Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd & Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Xiangqing Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd & Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Zifeng Wu
- Department of Anesthesiology, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, 214062, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|