1
|
Mafe AN, Büsselberg D. Could a Mediterranean Diet Modulate Alzheimer's Disease Progression? The Role of Gut Microbiota and Metabolite Signatures in Neurodegeneration. Foods 2025; 14:1559. [PMID: 40361641 PMCID: PMC12071848 DOI: 10.3390/foods14091559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), the most common form of dementia, represent a growing global health crisis, yet current treatment strategies remain primarily palliative. Recent studies have shown that neurodegeneration through complex interactions within the gut-brain axis largely depends on the gut microbiota and its metabolites. This review explores the intricate molecular mechanisms linking gut microbiota dysbiosis to cognitive decline, emphasizing the impact of microbial metabolites, including short-chain fatty acids (SCFAs), bile acids, and tryptophan metabolites, on neuroinflammation, blood-brain barrier (BBB) integrity, and amyloid-β and tau pathology. The paper highlights major microbiome signatures associated with Alzheimer's disease, detailing their metabolic pathways and inflammatory crosstalk. Dietary interventions have shown promise in modulating gut microbiota composition, potentially mitigating neurodegenerative processes. This review critically examines the influence of dietary patterns, such as the Mediterranean and Western diets, on microbiota-mediated neuroprotection. Bioactive compounds like prebiotics, omega-3 fatty acids, and polyphenols exhibit neuroprotective effects by modulating gut microbiota and reducing neuroinflammation. Furthermore, it discusses emerging microbiome-based therapeutic strategies, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), as potential interventions for slowing Alzheimer's progression. Despite these advances, several knowledge gaps remain, including interindividual variability in microbiome responses to dietary interventions and the need for large-scale, longitudinal studies. The study proposes an integrative, precision medicine approach, incorporating microbiome science into Alzheimer's treatment paradigms. Ultimately, cognizance of the gut-brain axis at a mechanistic level could unlock novel therapeutic avenues, offering a non-invasive, diet-based strategy for managing neurodegeneration and improving cognitive health.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Ar-Rayyan P.O. Box 22104, Qatar
| |
Collapse
|
2
|
Ponce-Lopez T. Peripheral Inflammation and Insulin Resistance: Their Impact on Blood-Brain Barrier Integrity and Glia Activation in Alzheimer's Disease. Int J Mol Sci 2025; 26:4209. [PMID: 40362446 PMCID: PMC12072112 DOI: 10.3390/ijms26094209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairment, and synaptic dysfunction. The accumulation of amyloid beta (Aβ) plaques and hyperphosphorylated tau protein leads to neuronal dysfunction, neuroinflammation, and glial cell activation. Emerging evidence suggests that peripheral insulin resistance and chronic inflammation, often associated with type 2 diabetes (T2D) and obesity, promote increased proinflammatory cytokines, oxidative stress, and immune cell infiltration. These conditions further damage the blood-brain barrier (BBB) integrity and promote neurotoxicity and chronic glial cell activation. This induces neuroinflammation and impaired neuronal insulin signaling, reducing glucose metabolism and exacerbating Aβ accumulation and tau hyperphosphorylation. Indeed, epidemiological studies have linked T2D and obesity with an increased risk of developing AD, reinforcing the connection between metabolic disorders and neurodegeneration. This review explores the relationships between peripheral insulin resistance, inflammation, and BBB dysfunction, highlighting their role in glial activation and the exacerbation of AD pathology.
Collapse
Affiliation(s)
- Teresa Ponce-Lopez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico
| |
Collapse
|
3
|
Beltran-Velasco AI, Clemente-Suárez VJ. Impact of Peripheral Inflammation on Blood-Brain Barrier Dysfunction and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:2440. [PMID: 40141084 PMCID: PMC11942216 DOI: 10.3390/ijms26062440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
The blood-brain barrier (BBB) is essential for maintaining brain homeostasis by regulating molecular exchange between the systemic circulation and the central nervous system. However, its dysfunction, often driven by peripheral inflammatory processes, has been increasingly linked to the development and progression of neurodegenerative diseases such as Alzheimer's and Parkinson's. Emerging evidence suggests that the gut-brain axis plays a key role in BBB integrity, with intestinal dysbiosis and chronic inflammation contributing to barrier disruption through immune and metabolic pathways. Furthermore, the selective vulnerability of specific brain regions to BBB dysfunction appears to be influenced by regional differences in vascularization, metabolic activity, and permeability, making certain areas more susceptible to neurodegenerative processes. This review explored the molecular mechanisms linking peripheral inflammation, gut microbiota, and BBB dysfunction, emphasizing their role in neurodegeneration. A comprehensive literature review was conducted using Web of Science, PubMed, Scopus, Wiley, ScienceDirect, and Medline, covering publications from 2015 to 2025. The findings highlight a complex interplay between gut microbiota-derived metabolites, immune signaling, and BBB permeability, underscoring the need for targeted interventions such as microbiome modulation, anti-inflammatory therapies, and advanced drug delivery systems. The heterogeneity of the BBB across different brain regions necessitates the development of region-specific therapeutic strategies. Despite advancements, critical knowledge gaps persist regarding the precise mechanisms underlying BBB dysfunction. Future research should leverage cutting-edge methodologies such as single-cell transcriptomics and organ-on-chip models to translate preclinical findings into effective clinical applications. Addressing these challenges will be crucial for developing personalized therapeutic approaches to mitigate the impact of BBB dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Isabel Beltran-Velasco
- NBC Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain;
| | - Vicente Javier Clemente-Suárez
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
4
|
Donaldson AIC, Fyfe CL, Martin JC, Smith EE, Horgan GW, Myint PK, Johnstone AM, Scott KP. Aging Gut-Brain Interactions: Pro-Inflammatory Gut Bacteria Are Elevated in Fecal Samples from Individuals Living with Alzheimer's Dementia. Geriatrics (Basel) 2025; 10:37. [PMID: 40126287 PMCID: PMC11932241 DOI: 10.3390/geriatrics10020037] [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: 11/29/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 03/25/2025] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is the most common form of dementia, characterized by an irreversible decline in cognitive function. The pathogenesis of several neurodegenerative disorders has been linked to changes in the gut microbiota, transmitted through the gut-brain axis. Methods: We set out to establish by case-control study methodology whether there were any differences in the composition and/or function of the gut microbiota between older resident adults in care homes with or without an AD diagnosis via analysis of the microbial composition from fecal samples. Results: The microbial composition, determined by 16S rRNA gene profiling, indicated that AD sufferers had significantly increased proportions of Escherichia/Shigella and Clostridium_sensu_stricto_1, and significantly decreased proportions of Bacteroides, Faecalibacterium, Blautia, and Roseburia species. The increase in potentially pro-inflammatory bacteria was consistent with slightly higher concentrations of calprotectin, a biomarker of gut inflammation. Fecal concentrations of most microbial metabolites measured were similar across groups, although participants with AD had significantly increased proportions of the branched-chain fatty acid, iso-butyrate, and lower overall concentrations of total short chain fatty acids. Conclusions: Participants with Alzheimer's disease have several key differences within their gut microbiota profile, in contrast to care home residents without Alzheimer's disease. The altered microbiome included both compositional and functional changes linked to poorer health and gut inflammation.
Collapse
Affiliation(s)
- Alison I. C. Donaldson
- Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK;
| | - Claire L. Fyfe
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK (A.M.J.); (K.P.S.)
| | - Jennifer C. Martin
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK (A.M.J.); (K.P.S.)
| | - Ellen E. Smith
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, 23 St Machar Drive, Aberdeen AB24 3FX, UK
| | - Graham W. Horgan
- Biomathematics and Statistics Scotland (BioSS), Aberdeen AB25 2ZD, UK
| | - Phyo K. Myint
- Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK;
| | - Alexandra M. Johnstone
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK (A.M.J.); (K.P.S.)
| | - Karen P. Scott
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK (A.M.J.); (K.P.S.)
| |
Collapse
|
5
|
Bibi A, Zhang F, Shen J, Din AU, Xu Y. Behavioral alterations in antibiotic-treated mice associated with gut microbiota dysbiosis: insights from 16S rRNA and metabolomics. Front Neurosci 2025; 19:1478304. [PMID: 40092066 PMCID: PMC11906700 DOI: 10.3389/fnins.2025.1478304] [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: 08/09/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The gut and brain interact through various metabolic and signaling pathways, each of which influences mental health. Gut dysbiosis caused by antibiotics is a well-known phenomenon that has serious implications for gut microbiota-brain interactions. Although antibiotics disrupt the gut microbiota's fundamental structure, the mechanisms that modulate the response and their impact on brain function are still unclear. It is imperative to comprehend and investigate crucial regulators and factors that play important roles. We aimed to study the effect of long-term antibiotic-induced disruption of gut microbiota, host metabolomes, and brain function and, particularly, to determine the basic interactions between them by treating the C57BL/6 mice with two different, most commonly used antibiotics, ciprofloxacin and amoxicillin. Anxiety-like behavior was confirmed by the elevated plus-maze test and open field test. Gut microbes and their metabolite profiles in fecal, serum, and brain samples were determined by 16S rRNA sequencing and untargeted metabolomics. In our study, long-term antibiotic treatment exerted anxiety-like effects. The fecal microbiota and metabolite status revealed that the top five genera found were Lactobacillus, Bacteroides, Akkermansia, Ruminococcus_gnavus_group, and unclassified norank_f_Muribaculaceae. The concentration of serotonin, L-Tyrosine, 5-Hydroxy-L-tryptophan, L-Glutamic acid, L-Glutamate, 5-Hydroxyindole acetic acid, and dopaminergic synapsis was comparatively low, while adenosine was high in antibiotic-treated mice. The KEGG enrichment analysis of serum and brain samples showed that amino acid metabolism pathways, such as tryptophan metabolism, threonine metabolism, serotonergic synapsis, methionine metabolism, and neuroactive ligand-receptor interaction, were significantly decreased in antibiotic-treated mice. Our study demonstrates that long-term antibiotic use induces gut dysbiosis and alters metabolic responses, leading to the dysregulation of brain signaling molecules and anxiety-like behavior. These findings highlight the complex interactions between gut microbiota and metabolic functions, providing new insights into the influence of microbial communities on gut-brain communication.
Collapse
Affiliation(s)
- Asma Bibi
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Famin Zhang
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jilong Shen
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ahmad Ud Din
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Yuanhong Xu
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Yi C, Huang S, Zhang W, Guo L, Xia T, Huang F, Yan Y, Li H, Yu B. Synergistic interactions between gut microbiota and short chain fatty acids: Pioneering therapeutic frontiers in chronic disease management. Microb Pathog 2025; 199:107231. [PMID: 39681288 DOI: 10.1016/j.micpath.2024.107231] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Microorganisms in the gut play a pivotal role in human health, influencing various pathophysiological processes. Certain microorganisms are particularly essential for maintaining intestinal homeostasis, reducing inflammation, supporting nervous system function, and regulating metabolic processes. Short-chain fatty acids (SCFAs) are a subset of fatty acids produced by the gut microbiota (GM) during the fermentation of indigestible polysaccharides. The interaction between GM and SCFAs is inherently bidirectional: the GM not only shapes SCFAs composition and metabolism but SCFAs also modulate microbiota's diversity, stability, growth, proliferation, and metabolism. Recent research has shown that GM and SCFAs communicate through various pathways, mainly involving mechanisms related to inflammation and immune responses, intestinal barrier function, the gut-brain axis, and metabolic regulation. An imbalance in GM and SCFA homeostasis can lead to the development of several chronic diseases, including inflammatory bowel disease, colorectal cancer, systemic lupus erythematosus, Alzheimer's disease, and type 2 diabetes mellitus. This review explores the synergistic interactions between GM and SCFAs, and how these interactions directly or indirectly influence the onset and progression of various diseases through the regulation of the mechanisms mentioned above.
Collapse
Affiliation(s)
- Chunmei Yi
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shanshan Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenlan Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Guo
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tong Xia
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fayin Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yijing Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Huhu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Bin Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
7
|
Abdelhamid M, Counts SE, Zhou C, Hida H, Kim JI, Michikawa M, Jung CG. Protective Effects of Bifidobacterium Breve MCC1274 as a Novel Therapy for Alzheimer's Disease. Nutrients 2025; 17:558. [PMID: 39940416 PMCID: PMC11820889 DOI: 10.3390/nu17030558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by memory impairment that significantly interferes with daily life. Therapeutic options for AD that substantively modify disease progression remain a critical unmet need. In this regard, the gut microbiota is crucial in maintaining human health by regulating metabolism and immune responses, and increasing evidence suggests that probiotics, particularly beneficial bacteria, can enhance memory and cognitive functions. Recent studies have highlighted the positive effects of Bifidobacterium breve MCC1274 (B. breve MCC1274) on individuals with mild cognitive impairment (MCI) and schizophrenia. Additionally, oral supplementation with B. breve MCC1274 has been shown to effectively prevent memory decline in AppNL-G-F mice. In relation to Alzheimer's pathology, oral supplementation with B. breve MCC1274 has been found to reduce amyloid-β (Aβ) accumulation and tau phosphorylation in both AppNL-G-F and wild-type (WT) mice. It also decreases microglial activation and increases levels of synaptic proteins. In this review, we examine the beneficial effects of B. breve MCC1274 on AD, exploring potential mechanisms of action and how this probiotic strain may aid in preventing or treating the disease. Furthermore, we discuss the broader implications of B. breve MCC1274 for improving overall host health and provide insights into future research directions for this promising probiotic therapy.
Collapse
Affiliation(s)
- Mona Abdelhamid
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503, USA; (M.A.); (S.E.C.)
| | - Scott E. Counts
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503, USA; (M.A.); (S.E.C.)
| | - Chunyu Zhou
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Jae-Il Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Makoto Michikawa
- Department of Geriatric Medicine, School of Life Dentistry at Niigata, Nippon Dental University, Niigata 951-8580, Japan
| | - Cha-Gyun Jung
- Center for Nursing International Promotion, Nagoya City University Graduate School of Nursing, Nagoya 467-8601, Japan
| |
Collapse
|
8
|
Dhanawat M, Malik G, Wilson K, Gupta S, Gupta N, Sardana S. The Gut Microbiota-Brain Axis: A New Frontier in Alzheimer's Disease Pathology. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:7-20. [PMID: 38967078 DOI: 10.2174/0118715273302508240613114103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Dr. Aloysius Alzheimer, a German neuropathologist and psychiatrist, recognized the primary instance of Alzheimer's disease (AD) for a millennium, and this ailment, along with its related dementias, remains a severe overall community issue related to health. Nearly fifty million individuals worldwide suffer from dementia, with Alzheimer's illness contributing to between 60 and 70% of the instances, estimated through the World Health Organization. In addition, 82 million individuals are anticipated to be affected by the global dementia epidemic by 2030 and 152 million by 2050. Furthermore, age, environmental circumstances, and inherited variables all increase the likelihood of acquiring neurodegenerative illnesses. Most recent pharmacological treatments are found in original hypotheses of disease, which include cholinergic (drugs that show affective cholinergic system availability) as well as amyloid-accumulation (a single drug is an antagonist receptor of Nmethyl D-aspartate). In 2020, the FDA provided approval on anti-amyloid drugs. According to mounting scientific data, this gut microbiota affects healthy physiological homeostasis and has a role in the etiology of conditions that range between obesity and neurodegenerative disorders like Alzheimer's. The microbiota-gut-brain axis might facilitate interconnection among gut microbes as well as the central nervous system (CNS). Interaction among the microbiota-gut system as well as the brain occurs through the "two-way" microbiota-gut-brain axis. Along this axis, the stomach as well as the brain develop physiologically and take on their final forms. This contact is constant and is mediated by numerous microbiota-derived products. The gut microbiota, for instance, can act as non-genetic markers to set a threshold for maintaining homeostasis or getting ill. The scientific community has conducted research and found that bowel dysbiosis and gastrointestinal tract dysregulation frequently occur in Alzheimer's disease (AD) patients. In this review, the effects of the microbiota- gut-brain axis on AD pathogenesis will be discussed.
Collapse
Affiliation(s)
- Meenakshi Dhanawat
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| | - Garima Malik
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Kashish Wilson
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Sumeet Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Nidhi Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Satish Sardana
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| |
Collapse
|
9
|
Cui J, Xiao S, Cao Y, Zhang Y, Yang J, Zheng L, Zhao F, Liu X, Liu D, Zhou Z, Wang P. Organophosphate Insecticide Malathion Induces Alzheimer's Disease-Like Cognitive Impairment in Mice: Evidence of the Microbiota-Gut-Brain Axis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:21966-21977. [PMID: 39545844 DOI: 10.1021/acs.est.4c07427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Evidence suggests that exposure to organophosphate pesticides increases the risk of neurodegenerative diseases, but the mechanisms remain unclear. This study investigated the effects of malathion on Alzheimer's disease (AD)-like symptoms at environmentally relevant concentrations using wild-type (WT) and APP/PS1 transgenic mouse models. Results showed that malathion exposure induced AD-like cognitive impairment, amyloid-β (Aβ) accumulation, and neuroinflammation in WT mice, with worsened symptoms in APP/PS1 mice. Mechanistic studies revealed that malathion induced AD-like gut microbiota dysbiosis (reduced Lactobacillus and Akkermansia, and increased Dubosiella), causing gut barrier impairment and tryptophan metabolism disruptions. This resulted in a significant increase in indole derivatives and activation of the colonic aryl hydrocarbon receptor (AhR), promoting the kynurenine (KYN) pathway while inhibiting the serotonin (5-HT) pathway. Increased neurotoxic KYN metabolites (3-hydroxykynurenine and quinolinic acid) triggered gut and systemic inflammation, upregulating hippocampal IL-6 and IL-1β mRNA levels and thereby causing neuroinflammation. Gut tryptophan metabolism disruptions caused hippocampal neurotransmitter imbalances, reducing the levels of 5-HT and its derivatives. These effects promoted AD progression in both WT and APP/PS1 mice. This study highlights the crucial role of the microbiota-gut-brain axis in AD-like cognitive impairment induced by malathion exposure, providing insights into the neurodegenerative disease risks posed by organophosphate pesticides.
Collapse
Affiliation(s)
- Jingna Cui
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Shouchun Xiao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yue Cao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Yaru Zhang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Jiaxing Yang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Li Zheng
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Fanrong Zhao
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Xueke Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Donghui Liu
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| | - Peng Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P. R. China
| |
Collapse
|
10
|
Pasupalak JK, Rajput P, Gupta GL. Gut microbiota and Alzheimer's disease: Exploring natural product intervention and the Gut-Brain axis for therapeutic strategies. Eur J Pharmacol 2024; 984:177022. [PMID: 39362390 DOI: 10.1016/j.ejphar.2024.177022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Numerous studies conducted over the last ten years have shown a strong correlation between the gut microbiota and the onset and progression of Alzheimer's disease (AD). However, the exact underlying mechanism is still unknown. An ongoing communication mechanism linking the gut and the brain is highlighted by the term "microbiota-gut-brain axis," which was originally coined the "gut-brain axis." Key metabolic, endocrine, neurological, and immunological mechanisms are involved in the microbiota‒gut‒brain axis and are essential for preserving brain homeostasis. Thus, the main emphasis of this review is how the gut microbiota contributes to the development of AD and how various natural products intervene in this disease. The first part of the review provides an outline of various pathways and relationships between the brain and gut microbiota, and the second part provides various mechanisms involved in the gut microbiota and AD. Finally, this review provides knowledge about natural products and their effectiveness in treating gut microbiota-induced AD. AD may be treated in the future by altering the gut microbiota with a customized diet, probiotics/prebiotics, plant products, and natural products. This entails altering the microbiological partners and products (such as amyloid protein) that these partners generate.
Collapse
Affiliation(s)
- Jajati K Pasupalak
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Prabha Rajput
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Girdhari Lal Gupta
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
11
|
Zeng M, Peng M, Liang J, Sun H. The Role of Gut Microbiota in Blood-Brain Barrier Disruption after Stroke. Mol Neurobiol 2024; 61:9735-9755. [PMID: 37498481 DOI: 10.1007/s12035-023-03512-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Growing evidence has proved that alterations in the gut microbiota have been linked to neurological disorders including stroke. Structural and functional disruption of the blood-brain barrier (BBB) is observed after stroke. In this context, there is pioneering evidence supporting that gut microbiota may be involved in the pathogenesis of stroke by regulating the BBB function. However, only a few experimental studies have been performed on stroke models to observe the BBB by altering the structure of gut microbiota, which warrant further exploration. Therefore, in order to provide a novel mechanism for stroke and highlight new insights into BBB modification as a stroke intervention, this review summarizes existing evidence of the relationship between gut microbiota and BBB integrity and discusses the mechanisms of gut microbiota on BBB dysfunction and its role in stroke.
Collapse
Affiliation(s)
- Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Yan Y, Gao Y, Kumar G, Fang Q, Yan H, Zhang N, Zhang Y, Song L, Li J, Zheng Y, Zhang N, Zhang P, Ma C. Exosomal MicroRNAs modulate the cognitive function in fasudil treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice model of Alzheimer's disease. Metab Brain Dis 2024; 39:1335-1351. [PMID: 39088109 PMCID: PMC11513711 DOI: 10.1007/s11011-024-01395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline stemming from the accumulation of beta-amyloid (Aβ) plaques and the propagation of tau pathology through synapses. Exosomes, crucial mediators in neuronal development, maintenance, and intercellular communication, have gained attention in AD research. Yet, the molecular mechanisms involving exosomal miRNAs in AD remain elusive. In this study, we treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice, a model for AD, with either vehicle (ADNS) or fasudil (ADF), while C57BL/6 (control) mice received vehicle (WT). Cognitive function was evaluated using the Y-maze test, and AD pathology was confirmed through immunostaining and western blot analysis of Aβ plaques and phosphorylated tau. Exosomal RNAs were extracted, sequenced, and analyzed from each mouse group. Our findings revealed that fasudil treatment improved cognitive function in AD mice, as evidenced by increased spontaneous alternation in the Y-maze test and reduced Aβ plaque load and phosphorylated tau protein expression in the hippocampus. Analysis of exosomal miRNAs identified three miRNAs (mmu-let-7i-5p, mmu-miR-19a-3p, mmu-miR-451a) common to both ADNS vs ADF and WT vs ADNS groups. Utilizing miRTarBase software, we predicted and analyzed target genes associated with these miRNAs. Gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of miRNA target genes indicated that mmu-miR-19a-3p and mmu-miR-451a are implicated in signal transduction, immune response, cellular communication, and nervous system pathways. Specifically, mmu-miR-19a-3p targeted genes involved in the sphingolipid signaling pathway, such as Pten and Tnf, while mmu-miR-451a targeted Nsmaf, Gnai3, and Akt3. Moreover, mmu-miR-451a targeted Myc in signaling pathways regulating the pluripotency of stem cells. In conclusion, fasudil treatment enhanced cognitive function by modulating exosomal MicroRNAs, particularly mmu-miR-451a and mmu-miR-19a-3p. These miRNAs hold promise as potential biomarkers and therapeutic targets for novel AD treatments.
Collapse
Affiliation(s)
- Yuqing Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
| | - Ye Gao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong.
| | - Qingli Fang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Hailong Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nianping Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yuna Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiehui Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yucheng Zheng
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nan Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Peijun Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Cungen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China.
| |
Collapse
|
13
|
Wu M, Cheng Y, Zhang R, Han W, Jiang H, Bi C, Zhang Z, Ye M, Lin X, Liu Z. Molecular mechanism and therapeutic strategy of bile acids in Alzheimer's disease from the emerging perspective of the microbiota-gut-brain axis. Biomed Pharmacother 2024; 178:117228. [PMID: 39088965 DOI: 10.1016/j.biopha.2024.117228] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/03/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-β outside neurons and Tau protein inside neurons. Various pathological mechanisms are implicated in AD, including brain insulin resistance, neuroinflammation, and endocrinal dysregulation of adrenal corticosteroids. These factors collectively contribute to neuronal damage and destruction. Recently, bile acids (BAs), which are metabolites of cholesterol, have shown neuroprotective potential against AD by targeting the above pathological changes. BAs can enter the systematic circulation and cross the blood-brain barrier, subsequently exerting neuroprotective effects by targeting several endogenous receptors. Additionally, BAs interact with the microbiota-gut-brain (MGB) axis to improve immune and neuroendocrine function during AD episodes. Gut microbes impact BA signaling in the brain through their involvement in BA biotransformation. In this review, we summarize the role and molecular mechanisms of BAs in AD while considering the MGB axis and propose novel strategies for preventing the onset and progression of AD.
Collapse
Affiliation(s)
- Menglu Wu
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China; Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yongyi Cheng
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ruolin Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Wenwen Han
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hanqi Jiang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Chenchen Bi
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ziyi Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Mengfei Ye
- Department of Psychiatry, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China
| | - Xiuqin Lin
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China.
| | - Zheng Liu
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China; Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
14
|
Jia R, Solé-Guardia G, Kiliaan AJ. Blood-brain barrier pathology in cerebral small vessel disease. Neural Regen Res 2024; 19:1233-1240. [PMID: 37905869 PMCID: PMC11467932 DOI: 10.4103/1673-5374.385864] [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: 05/31/2023] [Revised: 07/27/2023] [Accepted: 08/22/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cerebral small vessel disease is a neurological disease that affects the brain microvasculature and which is commonly observed among the elderly. Although at first it was considered innocuous, small vessel disease is nowadays regarded as one of the major vascular causes of dementia. Radiological signs of small vessel disease include small subcortical infarcts, white matter magnetic resonance imaging hyperintensities, lacunes, enlarged perivascular spaces, cerebral microbleeds, and brain atrophy; however, great heterogeneity in clinical symptoms is observed in small vessel disease patients. The pathophysiology of these lesions has been linked to multiple processes, such as hypoperfusion, defective cerebrovascular reactivity, and blood-brain barrier dysfunction. Notably, studies on small vessel disease suggest that blood-brain barrier dysfunction is among the earliest mechanisms in small vessel disease and might contribute to the development of the hallmarks of small vessel disease. Therefore, the purpose of this review is to provide a new foundation in the study of small vessel disease pathology. First, we discuss the main structural domains and functions of the blood-brain barrier. Secondly, we review the most recent evidence on blood-brain barrier dysfunction linked to small vessel disease. Finally, we conclude with a discussion on future perspectives and propose potential treatment targets and interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| |
Collapse
|
15
|
Joseph CR. Progressive Age-Associated Blood-Brain Barrier Leak/Dysfunction-Nexus of Neurodegenerative Disease Using MRI Markers to Identify Preclinical Disease and Potential New Targets for Future Treatments. Diagnostics (Basel) 2024; 14:726. [PMID: 38611639 PMCID: PMC11011559 DOI: 10.3390/diagnostics14070726] [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: 01/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
This review article focuses on the upstream pertinent pathophysiology leading to neurodegenerative disease. Specifically, the nexus appears to be blood-brain barrier (BBB) leakiness resulting in a two-prong inflammatory disease spectrum damaging the microvasculature and corrupting protein synthesis and degradation with accumulating misfolded toxic proteins. The suboptimal results of removing misfolded proteins mean a new approach to disease in the preclinical state is required aimed at other targets. Validated noninvasive imaging and serologic biomarkers of early preclinical disease implemented in the high-risk patient cohort along with periodic surveillance once effective treatments are developed will be required. This review discusses the physiology and pathophysiology of the BBB, new MRI imaging techniques identifying the leak, and altered fluid dynamic effects in the preclinical state. The risk factors for disease development, preventative measures, and potential treatment targets are also discussed.
Collapse
Affiliation(s)
- Charles R Joseph
- Neurology and Internal Medicine, College of Osteopathic Medicine, Liberty University, Lynchburg, VA 24502, USA
| |
Collapse
|
16
|
Wang S, Luo J, Wang H, Chen T, Sun J, Xi Q, Zhang Y. Extracellular Vesicles: A Crucial Player in the Intestinal Microenvironment and Beyond. Int J Mol Sci 2024; 25:3478. [PMID: 38542448 PMCID: PMC10970531 DOI: 10.3390/ijms25063478] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The intestinal ecological environment plays a crucial role in nutrient absorption and overall well-being. In recent years, research has focused on the effects of extracellular vesicles (EVs) in both physiological and pathological conditions of the intestine. The intestine does not only consume EVs from exogenous foods, but also those from other endogenous tissues and cells, and even from the gut microbiota. The alteration of conditions in the intestine and the intestinal microbiota subsequently gives rise to changes in other organs and systems, including the central nervous system (CNS), namely the microbiome-gut-brain axis, which also exhibits a significant involvement of EVs. This review first gives an overview of the generation and isolation techniques of EVs, and then mainly focuses on elucidating the functions of EVs derived from various origins on the intestine and the intestinal microenvironment, as well as the impacts of an altered intestinal microenvironment on other physiological systems. Lastly, we discuss the role of microbial and cellular EVs in the microbiome-gut-brain axis. This review enhances the understanding of the specific roles of EVs in the gut microenvironment and the central nervous system, thereby promoting more effective treatment strategies for certain associated diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.W.); (J.L.); (H.W.); (T.C.); (J.S.); (Q.X.)
| |
Collapse
|
17
|
Bu L, Wang C, Bai J, Song J, Zhang Y, Chen H, Suo H. Gut microbiome-based therapies for alleviating cognitive impairment: state of the field, limitations, and future perspectives. Food Funct 2024; 15:1116-1134. [PMID: 38224464 DOI: 10.1039/d3fo02307a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Cognitive impairment (CI) is a multifaceted neurological condition that can trigger negative emotions and a range of concurrent symptoms, imposing significant public health and economic burdens on society. Therefore, it is imperative to discover a remedy for CI. Nevertheless, the mechanisms behind the onset of this disease are multifactorial, which makes the search for effective amelioration difficult and complex, hindering the search for effective measures. Intriguingly, preclinical research indicates that gut microbiota by influencing brain function, plays an important role in the progression of CI. Furthermore, numerous preclinical studies have highlighted the potential of probiotics, prebiotics, fecal microbiota transplantation (FMT), and diet in modulating the gut microbiota, thereby ameliorating CI symptoms. This review provides a comprehensive evaluation of CI pathogenesis, emphasizing the contribution of gut microbiota disorders to CI development. It also summarizes and discusses current strategies and mechanisms centered on the synergistic role of gut microbiota modulation in the microbiota-gut-brain axis in CI development. Finally, problems with existing approaches are contemplated and the development of microbial modulation strategies as therapeutic approaches to promote and restore brain cognition is discussed. Further research considerations and directions are highlighted to provide ideas for future CI prevention and treatment strategies.
Collapse
Affiliation(s)
- Linli Bu
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400715, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Yuhong Zhang
- Institute of Food Sciences and Technology, Tibet Academy of Agricultural and Animal Husbandry Sciences, Xizang 850000, China
| | - Hongyu Chen
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| |
Collapse
|
18
|
Yeske B, Hou J, Chu DY, Adluru N, Nair VA, Beniwal-Patel P, Saha S, Prabhakaran V. Structural brain morphometry differences and similarities between young patients with Crohn's disease in remission and healthy young and old controls. Front Neurosci 2024; 18:1210939. [PMID: 38356645 PMCID: PMC10864509 DOI: 10.3389/fnins.2024.1210939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Crohn's disease (CD), one of the main phenotypes of inflammatory bowel disease (IBD), can affect any part of the gastrointestinal tract. It can impact the function of gastrointestinal secretions, as well as increasing the intestinal permeability leading to an aberrant immunological response and subsequent intestinal inflammation. Studies have reported anatomical and functional brain changes in Crohn's Disease patients (CDs), possibly due to increased inflammatory markers and microglial cells that play key roles in communicating between the brain, gut, and systemic immune system. To date, no studies have demonstrated similarities between morphological brain changes seen in IBD and brain morphometry observed in older healthy controls.. Methods For the present study, twelve young CDs in remission (M = 26.08 years, SD = 4.9 years, 7 male) were recruited from an IBD Clinic. Data from 12 young age-matched healthy controls (HCs) (24.5 years, SD = 3.6 years, 8 male) and 12 older HCs (59 years, SD = 8 years, 8 male), previously collected for a different study under a similar MR protocol, were analyzed as controls. T1 weighted images and structural image processing techniques were used to extract surface-based brain measures, to test our hypothesis that young CDs have different brain surface morphometry than their age-matched young HCs and furthermore, appear more similar to older HCs. The phonemic verbal fluency (VF) task (the Controlled Oral Word Association Test, COWAT) (Benton, 1976) was administered to test verbal cognitive ability and executive control. Results/Discussion On the whole, CDs had more brain regions with differences in brain morphometry measures when compared to the young HCs as compared to the old HCs, suggesting that CD has an effect on the brain that makes it appear more similar to old HCs. Additionally, our study demonstrates this atypical brain morphometry is associated with function on a cognitive task. These results suggest that even younger CDs may be showing some evidence of structural brain changes that demonstrate increased resemblance to older HC brains rather than their similarly aged healthy counterparts.
Collapse
Affiliation(s)
- Benjamin Yeske
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Jiancheng Hou
- Center for Cross-Straits Cultural Development, Fujian Normal University, Fuzhou City, Fujian, China
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Daniel Y. Chu
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Nagesh Adluru
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- The Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Veena A. Nair
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Poonam Beniwal-Patel
- Gastroenterology and Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sumona Saha
- Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin- Madison, Madison, WI, United States
| | - Vivek Prabhakaran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Psychology and Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
19
|
Bradley E, Haran J. The human gut microbiome and aging. Gut Microbes 2024; 16:2359677. [PMID: 38831607 PMCID: PMC11152108 DOI: 10.1080/19490976.2024.2359677] [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: 01/19/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The composition of the human gut microbiome has been observed to change over the course of an individual's life. From birth, it is shaped by mode of delivery, diet, environmental exposures, geographic location, exposures to medications, and by aging itself. Here, we present a narrative review of the gut microbiome across the lifespan with a focus on its impacts on aging and age-related diseases in humans. We will describe how it is shaped, and features of the gut microbiome that have been associated with diseases at different phases of life and how this can adversely affect healthy aging. Across the lifespan, and especially in old age, a diverse microbiome that includes organisms suspected to produce anti-inflammatory metabolites such as short-chain fatty acids, has been reported to be associated with healthy aging. These findings have been remarkably consistent across geographic regions of the world suggesting that they could be universal features of healthy aging across all cultures and genetic backgrounds. Exactly how these features of the microbiome affect biologic processes associated with aging thus promoting healthy aging will be crucial to targeting the gut microbiome for interventions that will support health and longevity.
Collapse
Affiliation(s)
- Evan Bradley
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| | - John Haran
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| |
Collapse
|
20
|
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
|
21
|
Northuis CA, Bell EJ, Lutsey PL, George KM, Gottesman RF, Mosley TH, Whitsel EA, Lakshminarayan K. Cumulative Use of Proton Pump Inhibitors and Risk of Dementia: The Atherosclerosis Risk in Communities Study. Neurology 2023; 101:e1771-e1778. [PMID: 37558503 PMCID: PMC10634644 DOI: 10.1212/wnl.0000000000207747] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/20/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Studies on the association between proton pump inhibitor (PPI) use and dementia report mixed results and do not examine the impact of cumulative PPI use. We evaluated the associations between current and cumulative PPI use and risk of incident dementia in the Atherosclerosis Risk in Communities (ARIC) Study. METHODS These analyses used participants from a community-based cohort (ARIC) from the time of enrollment (1987-1989) through 2017. PPI use was assessed through visual medication inventory at clinic visits 1 (1987-1989) to 5 (2011-2013) and reported annually in study phone calls (2006-2011). This study uses ARIC visit 5 as baseline because this was the first visit in which PPI use was common. PPI use was examined 2 ways: current use at visit 5 and duration of use before visit 5 (from visit 1 to 2011, exposure categories: 0 day, 1 day-2.8 years, 2.8-4.4 years, >4.4 years). The outcome was incident dementia after visit 5. Cox proportional hazard models were used, adjusted for demographics, comorbid conditions, and other medication use. RESULTS A total of 5,712 dementia-free participants at visit 5 (mean age 75.4 ± 5.1 years; 22% Black race; 58% female) were included in our analysis. The median follow-up was 5.5 years. The minimum cumulative PPI use was 112 days, and the maximum use was 20.3 years. There were 585 cases of incident dementia identified during follow-up. Participants using PPIs at visit 5 were not at a significantly higher risk of developing dementia during subsequent follow-up than those not using PPIs (hazard ratio (HR): 1.1 [95% confidence interval (CI) 0.9-1.3]). Those who used PPIs for >4.4 cumulative years before visit 5 were at 33% higher risk of developing dementia during follow-up (HR: 1.3 [95% CI 1.0-1.8]) than those reporting no use. Associations were not significant for lesser durations of PPI use. DISCUSSION Future studies are needed to understand possible pathways between cumulative PPI use and the development of dementia. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that the use of prescribed PPIs for >4.4 years by individuals aged 45 years and older is associated with a higher incidence of newly diagnosed dementia.
Collapse
Affiliation(s)
- Carin A Northuis
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Elizabeth J Bell
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Pamela L Lutsey
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Kristen M George
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Rebecca F Gottesman
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Tom H Mosley
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Eric A Whitsel
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill
| | - Kamakshi Lakshminarayan
- From the University of Minnesota (C.A.N., P.L.L., K.L.), Minneapolis; Optum (E.J.B.), Minneapolis, MN; University of California, Davis (K.M.G.); National Institute of Neurological Disorders and Stroke (R.F.G.), Washington, DC; University of Mississippi Medical Center (T.H.M.), Jackson; and University of North Carolina (E.A.W.), Chapel Hill.
| |
Collapse
|
22
|
Li Z, Jiang Y, Long C, Peng Q, Yue R. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci Ther 2023; 29 Suppl 1:59-73. [PMID: 36601656 PMCID: PMC10314112 DOI: 10.1111/cns.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) is one of the most insidious complications of type 2 diabetes mellitus, which can seriously affect the ability to self-monitoring of blood glucose and the quality of life in the elderly. Previous pathological studies of cognitive dysfunction have focused on neuronal dysfunction, characterized by extracellular beta-amyloid deposition and intracellular tau hyperphosphorylation. In recent years, astrocytes have been recognized as a potential therapeutic target for cognitive dysfunction and important participants in the central control of metabolism. The disorder of gut microbiota and their metabolites have been linked to a series of metabolic diseases such as diabetes mellitus. The imbalance of intestinal flora has the effect of promoting the occurrence and deterioration of several diabetes-related complications. Gut microbes and their metabolites can drive astrocyte activation. AIMS We reviewed the pathological progress of DCD related to the "gut microbiota-astrocyte" axis in terms of peripheral and central inflammation, intestinal and blood-brain barrier (BBB) dysfunction, systemic and brain energy metabolism disorders to deepen the pathological research progress of DCD and explore the potential therapeutic targets. CONCLUSION "Gut microbiota-astrocyte" axis, unique bidirectional crosstalk in the brain-gut axis, mediates the intermediate pathological process of neurocognitive dysfunction secondary to metabolic disorders in diabetes mellitus.
Collapse
Affiliation(s)
- Zi‐Han Li
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ya‐Yi Jiang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Cai‐Yi Long
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Qian Peng
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ren‐Song Yue
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
23
|
Khezri MR, Ghasemnejad-Berenji M. Gut microbiota and circadian rhythm in Alzheimer's disease pathophysiology: a review and hypothesis on their association. NPJ AGING 2023; 9:9. [PMID: 37130863 PMCID: PMC10154390 DOI: 10.1038/s41514-023-00104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 03/15/2023] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and the leading cause of dementia worldwide. Different pathologic changes have been introduced to be involved in its progression. Although amyloid-β (Aβ) deposition and tau hyperphosphorylation and aggregation are mainly considered the main characterizations of AD, several other processes are involved. In recent years, several other changes, including alterations in gut microbiota proportion and circadian rhythms, have been noticed due to their role in AD progression. However, the exact mechanism indicating the association between circadian rhythms and gut microbiota abundance has not been investigated yet. This paper aims to review the role of gut microbiota and circadian rhythm in AD pathophysiology and introduces a hypothesis to explain their association.
Collapse
Affiliation(s)
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
- Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
24
|
Abstract
This narrative review seeks to examine the relationships between bacterial microbiomes and infectious disease. This is achieved by detailing how different human host microbiomes develop and function, from the earliest infant acquisitions of maternal and environmental species through to the full development of microbiomes by adulthood. Communication between bacterial species or communities of species within and outside of the microbiome is a factor in both maintenance of homeostasis and management of threats from the external environment. Dysbiosis of this homeostasis is key to understanding the development of disease states. Several microbiomes and the microbiota within are used as prime examples of how changes in species composition, particularly at the phylum level, leads to such diverse conditions as inflammatory bowel disease (IBD), type 2 diabetes, psoriasis, Parkinson's disease, reflux oesophagitis and others. The review examines spatial relationships between microbiomes to understand how dysbiosis in the gut microbiome in particular can influence diseases in distant host sites via routes such as the gut-lung, gut-skin and gut-brain axes. Microbiome interaction with host processes such as adaptive immunity is increasingly identified as critical to developing the capacity of the immune system to react to pathogens. Dysbiosis of essential bacteria involved in modification of host substrates such as bile acid components can result in development of Crohn's disease, small intestine bacterial overgrowth, hepatic cancer and obesity. Interactions between microbiomes in distantly located sites are being increasingly being identified, resulting in a 'whole of body' effect by the combined host microbiome.
Collapse
Affiliation(s)
- Jim Manos
- Infection, Immunity and InflammationSchool of Medical SciencesFaculty of Medicine and HealthThe Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| |
Collapse
|
25
|
Li ZL, Ma HT, Wang M, Qian YH. Research trend of microbiota-gut-brain axis in Alzheimer's disease based on CiteSpace (2012-2021): A bibliometrics analysis of 608 articles. Front Aging Neurosci 2022; 14:1036120. [PMID: 36483116 PMCID: PMC9724362 DOI: 10.3389/fnagi.2022.1036120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/03/2022] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Recently, research on the microbiota-gut-brain axis (MGBA) has received increasing attention, and the number of studies related to Alzheimer's disease (AD) has increased rapidly, but there is currently a lack of summary of MGBA in AD. OBJECTIVE To capture research hotspots, grasp the context of disciplinary research, and explore future research development directions. METHODS In the core dataset of Web of Science, documents are searched according to specific subject words. CiteSpace software is used to perform statistical analysis on measurement indicators such as the number of published papers, publishing countries, institutions, subject areas, authors, cocited journals, and keywords, and to visualize of a network of relevant content elements. RESULTS The research of MGBA in AD has shown an upward trend year by year, and the cooperation between countries is relatively close, and mainly involves the intersection of neuroscience, pharmacy, and microbiology. This research focuses on the relationship between MGBA and AD symptoms. Keyword hotspots are closely related to new technologies. Alzheimer's disease, anterior cingulate cortex, inflammatory degeneration, dysbiosis, and other research are the focus of this field. CONCLUSION The study revealed that the research and development of MGBA in AD rapidly progressed, but no breakthrough has been made in the past decade, it still needs to be closely combined with multidisciplinary technology to grasp the frontier hotspots. Countries should further strengthen cooperation, improve the disciplinary system, and increase the proportion of empirical research in all research.
Collapse
Affiliation(s)
- Zi-Long Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Hao-Tian Ma
- College of Forensic Science, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Meng Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Yi-Hua Qian
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
26
|
Tarawneh R, Penhos E. The gut microbiome and Alzheimer's disease: Complex and bidirectional interactions. Neurosci Biobehav Rev 2022; 141:104814. [PMID: 35934087 PMCID: PMC9637435 DOI: 10.1016/j.neubiorev.2022.104814] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022]
Abstract
Structural and functional alterations to the gut microbiome, referred to as gut dysbiosis, have emerged as potential key mediators of neurodegeneration and Alzheimer disease (AD) pathogenesis through the "gut -brain" axis. Emerging data from animal and clinical studies support an important role for gut dysbiosis in mediating neuroinflammation, central and peripheral immune dysregulation, abnormal brain protein aggregation, and impaired intestinal and brain barrier permeability, leading to neuronal loss and cognitive impairment. Gut dysbiosis has also been shown to directly influence various mechanisms involved in neuronal growth and repair, synaptic plasticity, and memory and learning functions. Aging and lifestyle factors including diet, exercise, sleep, and stress influence AD risk through gut dysbiosis. Furthermore, AD is associated with characteristic gut microbial signatures which offer value as potential markers of disease severity and progression. Together, these findings suggest the presence of a complex bidirectional relationship between AD and the gut microbiome and highlight the utility of gut modulation strategies as potential preventative or therapeutic strategies in AD. We here review the current literature regarding the role of the gut-brain axis in AD pathogenesis and its potential role as a future therapeutic target in AD treatment and/or prevention.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, Alzheimer Disease Research Center, The University of New Mexico, Albuquerque, NM 87106, USA.
| | - Elena Penhos
- College of Medicine, The Ohio State University, Columbus, OH, USA 43210
| |
Collapse
|
27
|
Gao J, Song G, Shen H, Wu Y, Zhao C, Zhang Z, Jiang Q, Li X, Ma X, Tan B, Yin Y. Allicin Improves Intestinal Epithelial Barrier Function and Prevents LPS-Induced Barrier Damages of Intestinal Epithelial Cell Monolayers. Front Immunol 2022; 13:847861. [PMID: 35185936 PMCID: PMC8854216 DOI: 10.3389/fimmu.2022.847861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
Gut barrier disruption is the initial pathogenesis of various diseases. We previously reported that dietary allicin improves tight junction proteins in the endoplasmic reticulum stressed jejunum. However, whether the allicin benefits the gut barrier within mycotoxin or endotoxin exposure is unknown. In the present study, IPEC-J2 cell monolayers within or without deoxynivalenol (DON) or lipopolysaccharide (LPS) challenges were employed to investigate the effects of allicin on intestinal barrier function and explore the potential mechanisms. Results clarified that allicin at 2 μg/mL increased the viability, whereas the allicin higher than 10 μg/mL lowered the viability of IPEC-J2 cells via inhibiting cell proliferation. Besides, allicin increased trans-epithelial electric resistance (TEER), decreased paracellular permeability, and enhanced ZO-1 integrity of the IPEC-J2 cell monolayers. Finally, allicin supplementation prevented the LPS-induced barrier damages via activating Nrf2/HO-1 pathway-dependent antioxidant system. In conclusion, the present study strongly confirmed allicin as an effective nutrient to improve intestinal barrier function and prevent bacterial endotoxin-induced barrier damages.
Collapse
Affiliation(s)
- Jingxia Gao
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Guanzhong Song
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Haibo Shen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yiming Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chongqi Zhao
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhuo Zhang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Qian Jiang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Qian Jiang,
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaokang Ma
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Bie Tan
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yulong Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|